US10172848B2 - Gap junction-enhancing agents for treatment of necrotizing enterocolitis and inflammatory bowel disease - Google Patents

Gap junction-enhancing agents for treatment of necrotizing enterocolitis and inflammatory bowel disease Download PDF

Info

Publication number
US10172848B2
US10172848B2 US13/921,865 US201313921865A US10172848B2 US 10172848 B2 US10172848 B2 US 10172848B2 US 201313921865 A US201313921865 A US 201313921865A US 10172848 B2 US10172848 B2 US 10172848B2
Authority
US
United States
Prior art keywords
gap junction
enhancing agent
seq
gap
nec
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
US13/921,865
Other versions
US20130345154A1 (en
Inventor
David J. Hackam
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of Pittsburgh
Original Assignee
University of Pittsburgh
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University of Pittsburgh filed Critical University of Pittsburgh
Priority to US13/921,865 priority Critical patent/US10172848B2/en
Publication of US20130345154A1 publication Critical patent/US20130345154A1/en
Assigned to UNIVERSITY OF PITTSBURGH - OF THE COMMONWEALTH SYSTEM OF HIGHER EDUCATION reassignment UNIVERSITY OF PITTSBURGH - OF THE COMMONWEALTH SYSTEM OF HIGHER EDUCATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HACKAM, DAVID J.
Priority to US16/194,009 priority patent/US20190070168A1/en
Application granted granted Critical
Publication of US10172848B2 publication Critical patent/US10172848B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/07Tetrapeptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/08Peptides having 5 to 11 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/10Peptides having 12 to 20 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]

Definitions

  • This application relates to agents, particularly peptides and peptide analogs, that enhance the functionality of gap junctions between intestinal enterocytes, and which may be used to treat disorders associated with impaired interenterocyte gap junctions such as necrotizing enterocolitis and inflammatory bowel diseases. These gap junction enhancing peptides may also be used to reduce the risk of occurrence of these disorders.
  • Necrotizing enterocolitis is a leading cause of death and disability in premature infants. Patients that develop NEC do so suddenly and without warning, and upon surgical exploration of the abdomen, frequently demonstrate large regions of the intestine that are either dead or dying. In over half the cases, patients that develop NEC do not survive their disease, and in survivors, an additional third will develop long term complications related to the initial development of the disease.
  • NEC Necrotizing enterocolitis
  • NEC is characterized by impaired enterocyte migration along the crypt-villus axis.
  • the impaired enterocyte migration results in persistent mucosal defects, bacterial translocation, and the development of systemic sepsis which leads to death in many cases.
  • enterocytes migrate together as sheets, and that enterocyte migration is dependent upon intact inter-enterocyte communication via Cx43-mediated gap junctions (1, 2).
  • the release of the pro-inflammatory cytokine interferon gamma (IFN ⁇ ) plays a critical role in the impairment of mucosal healing in part by inhibiting gap junctions between enterocytes (1, 2).
  • IBD human inflammatory bowel disease
  • Gap junctions are intercellular channels that exist between adjacent cells which allow the transfer of small molecules between adjoining cells.
  • Each gap junction channel is comprised of a pair of hexameric arrays of individual subunits called connexins, of which the most widely expressed isoform is connexin-43 (Cx43; 2, 3, 4).
  • the function of gap junctions is regulated in part through phosphorylation of the individual connexin molecules, which serves to regulate the localization of the channels at the plasma membrane as well as to regulate the channel through gating (5, 6).
  • the present invention relates to methods of reducing the risk of occurrence of, and/or treating, necrotizing enterocolitis (“NEC”) or inflammatory bowel disease (“IBD”) comprising administering, to a subject in need of such treatment, an effective amount of a gap junction enhancing agent (“GJEA”), for example a peptide (“GJP”) or peptide analog (“GRA”).
  • GJEA gap junction enhancing agent
  • GJP peptide
  • GAA peptide analog
  • FIG. 1 Cx43 knockout “cko” mice were generated. RT-PCR demonstrated the absence of Cx43.
  • FIG. 2A-B Immunohistochemistry studies using fluorescently labeled antibodies directed toward Cx43 and actin demonstrate (A) the presence of both proteins in wild-type intestinal villi and (B) the absence of Cx43 in the villi of knock-out animals having the selective deletion of Cx43.
  • FIG. 3A-C (A) Photomicrograph of intestinal villi in a wild-type mouse. (B) Photomicrograph of intestinal villi in a Cx43 knockout mouse. (C) migration rate of enterocytes in wild-type versus Cx43 knockout (“ ⁇ IEC”).
  • FIG. 4 Absorption of fat, protein and glucose by WT versus Cx43 knockout mouse intestine.
  • FIG. 5A-H (A) Photomicrograph of intestinal tissue of a healthy wild-type mouse; (B) photomicrograph of intestinal tissue of a healthy wild-type mouse stained to show expression of inducible nitric oxide synthase (“iNOS”); (C) photomicrograph of intestinal tissue of a wild-type mouse having NEC; (D) photomicrograph of intestinal tissue of a wild-type mouse having NEC stained to show expression of iNOS; (E) photomicrograph of intestinal tissue of a Cx43 knockout mouse; (F) photomicrograph of intestinal tissue of a Cx43 knockout mouse stained to show expression of iNOS; (G) photomicrograph of intestinal tissue of a Cx43 knockout mouse having NEC; and (H) photomicrograph of intestinal tissue of a Cx43 knockout mouse with NEC stained to show expression of iNOS.
  • iNOS inducible nitric oxide synthase
  • FIG. 6A-B (A) Migration rate of enterocytes from either wild-type (“WT”) healthy (control) mice, wild-type mice having NEC, Cx43 knockout mice (“ ⁇ IEC”), or Cx43 mice having NEC. (B) Cytokine levels of either wild-type or Cx43 knockout mice (“Cx43 ⁇ IEC ”), having NEC.
  • FIG. 7A-B Cx43 expression in (A) premature (gestation day 15.5) or (B) postnatal (day 10) murine intestinal cells, as shown using a fluorescent anti-Cx43 antibody.
  • FIG. 8A-E (A) Photomicrograph of control murine intestine.
  • B Photomicrograph of murine intestine after exposure to adenoviral vector carrying Green Fluorescent Protein (“GFP”) gene.
  • C Photomicrograph of murine intestine after exposure to adenoviral vector carrying GFP and dominant negative Cx43 mutant protein (“dnCx43”).
  • D Expression of GFP in murine intestine in the presence (“DN”) or absence (“WT”) of dnCx43.
  • E Colitis severity in murine intestine in the presence (“DN”) or absence (“WT”) of dnCx43.
  • FIG. 9A-C Surface mucosa of murine intestine, either (A) control; (B) after exposure to adenoviral vector carrying GFP; or (C) after exposure to adenoviral vector carrying GFP and dnCx43.
  • FIG. 10A-B IEC-6 enterocytes were treated with the concentrations of phorbol myristate acetate (“PMA”) indicated for one hour and then evaluated for the extent of enterocyte gap junction communication by confocal based dye transfer. The untreated cells represent the 100% dye transfer. *p ⁇ 0.05 versus untreated cells.
  • IEC-6 cells were treated with the concentration of PMA indicated and assessed for their ability to migrate into a scraped wound over 20 hours. *p ⁇ 0.05 vs. untreated cells. Representative of two separate experiments.
  • PMA phorbol myristate acetate
  • Gap junction enhancing agents include gap junction enhancing peptides (“GJPs”) and peptide analogs (“GJPAs”) as well as other compounds such as pharmacologic agents.
  • Non-limiting examples of pharmacologic agents that are GJEAs include phorbol myristate acetate (“PMA”) and quinoline derivatives as described in United States Patent Publication No. 20090143425, such as, but not limited to, primaquine, mefloquine, PQ2, PQ3, PQ4, PQ5, PQ6, PQ7, PQ8 and/or the compound PQ1:
  • PMA phorbol myristate acetate
  • Gap Junction enhancing peptides that may be used according to the invention include but are not limited to peptides listed in United States Patent Application Publication No. 20090075291 by Delmar et al., which is incorporated by reference in its entirety herein, including: DVPGRDPGYIKGGGSAHARVPFYSHSLNRNRKPSLYQ (SEQ ID NO:1); EIQPRSPLMFSGGGSAHARVPFYSHSAKEARWPRAHR (SEQ ID NO:2); GIAAREPNSHDGGGSAHARVPFYSHSRDLWRKPAKSL (SEQ ID NO:3); WEEPRRPFTMSGGGSAETHARVPFYSHSPMRHRLPGVHL (SEQ ID NO:4); SDDLRSPQLHNGGGSAVPFYSHSHMVRRKPRNPR (SEQ ID NO:5); GHLHLRVPTLKM (SEQ ID NO:6); EFIRSPHSVDWL (SEQ ID NO:7); SQSRNPPMPPPR (SEQ ID NO:1)
  • GJPA is Compound ZP123 (Rotigaptide (2R, 4S)-1-[(2R)-1-[(2R)-2-acetamido-3-(4-hydroxyphenyl)propanoyl]pyrrolidine-2-carbonyl]-N-[2-[[(2R)-1-[(2-amino-2-oxoethyl)amino]-1-oxopropan-2-yl]amino]-2-oxoethyl]-4-hydroxypyrrolidine-2-carboxamide), having the following formula:
  • Rotigaptide which enhances gap junction connectivity through increased activation of Cx43.
  • the prototypical compound, Rotigaptide is stable, has a long half-life, and is in clinical trials as an anti-arrhythmic agent in adults, based upon its documented ability to enhance gap junction connectivity.
  • GJPs include pharmacophores of Cx43 based upon the “RXP” series of Cx43-binding peptides. These pharmacophores bind to the carboxyl terminal of Cx43, and share a 34-aa peptide (RXP-E (SEQ ID NO:27)) sequence. Two representatives of this family are a cyclized heptapeptide (called CyRP-71) and a linear octapeptide of sequence RRNYRRNY (SEQ ID NO:28).
  • GJP Gly-Ala-Gly-Hyp-Pro-Tyr-NH 2
  • AAP10 Gly-Ala-Gly-Hyp-Pro-Tyr-NH 2
  • AAP10 Gly-Ala-Gly-Hyp-Pro-Tyr-NH 2
  • related sequences such as but not limited to Gly-Pro-Hyp-Gly-Ala-Gly (SEQ ID NO:30) or cyclo[CF(3)C(OH)-Gly-Ala-Gly-Hyp-Pro-Tyr (SEQ ID NO:31).
  • the present invention further provides for peptides that are between 4 and 100, or between 4 and 50, or between 6 and 100, or between 6 and 50, or between 8 and 100, or between 8 and 50, or between 10 and 100, or between 10 and 50, or between 15 and 35, amino acids long and comprise one or more peptide having SEQ ID NO:1-31.
  • the present invention further provides for peptides that are between 4 and 100, or between 4 and 50, or between 6 and 100, or between 6 and 50, or between 8 and 100, or between 8 and 50, or between 10 and 100, or between 10 and 50, or between 15 and 35, amino acids long and comprise one or more peptide having a sequence which differs from any of SEQ ID NO:1-31 by no more than one amino acid and has a gap junction enhancing activity that is at least 80 percent of a peptide lacking the difference in sequence.
  • GJPA peptide analogs
  • GAP-134 i.e. (2S,4R)-1-(2-aminoacetyl)-4-benzamido-pyrrolidine-2-carboxylic acid
  • GAP-134 acts by enhancing gap junction conductance without effects on other ion channels, without any apparent changes in transcription or distribution of Cx43.
  • This compound has a shorter half-life than ZP123, which may decrease its effectiveness, but also may limit potential toxicity;
  • GJEA, GJP or GJPA ability of a GJEA, GJP or GJPA to enhance gap junction communication between enterocytes may be determined by any method known in the art, including in vitro and in vivo studies.
  • gap junction intercellular communication (GJIC) studies which assess the ability of a GJEA, GJP or GJPA to enhance gap junction communication between enterocytes, may be performed in vitro in cultured enterocytes, using live cell confocal microscopy and fluorescence recovery after photobleaching (FRAP), as in (1, 2).
  • FRAP live cell confocal microscopy and fluorescence recovery after photobleaching
  • cells may be plated to confluence, and treated with a GJEA, GJP or GJPA for times between 1-4 hours, and the degree of gap junction intercellular communication may be determined using confocal based fluorescence recovery after photobleaching (FRAP), which measures the movement of a fluorescent tracer through gap junctions into an area that has been previously photobleached using a laser, such that the rate and extent to which the photobleached cells fill with the fluorescent dye (termed the “fluorescence recovery”) may provide a direct measure of gap junction activity.
  • FRAP confocal based fluorescence recovery after photobleaching
  • a second non-limiting example of a method for assessing the ability of a GJEA, GJP or GJPA to enhance gap junction communication between enterocytes is single cell microinjection (2), which allows the detection of the extent to which a detectable tracer, for example the 0.4 kilodalton fluorescent gap junction tracer Lucifer yellow, passes from an injected cell to adjacent cells through gap junctions.
  • a detectable tracer for example the 0.4 kilodalton fluorescent gap junction tracer Lucifer yellow
  • IEC-6 cells may be used for such studies, as they represent a well validated culture model of the intestine.
  • Non-limiting examples of other cell lines that may be used include HT-29 and CaCO-2 cells.
  • the ability of a GJEA, GJP or GJPA to treat NEC may be assessed in vivo by the following study in mice.
  • the ability of a GJEA, GJP or GJPA to treat IBD may be assessed in vivo by the following study in mice.
  • the foregoing methods may be used to determine whether a particular concentration of a GJEA, GJP or GJPA is able to enhance gap junction communication between enterocytes, to reduce the incidence of NEC, to treat NEC, to reduce the incidence of IBD, or to treat IBD.
  • aGJEA, GJP or GJPA enhances (increases the rate or amount of) communication between adjacent enterocytes by at least about 10 percent, or at least about 20 percent, or at least about 30 percent, or at least about 40 percent, or at least about 50 percent, or at least about 60 percent, or at least about 70 percent, or at least about 80 percent, or at least about 100 percent, relative to the amount of communication under control conditions (e.g. the absence of the GJEA, GJP or GJPA).
  • the present invention provides for a pharmaceutical composition
  • a pharmaceutical composition comprising one or more GJEA, GJP or GJPA in a pharmaceutically suitable carrier.
  • Said pharmaceutical composition may be in solid or liquid form.
  • a pharmaceutical composition may comprise GJEA, GJP or GJPA which is optionally lyophilized or comprised in micelles or microspheres.
  • a pharmaceutical composition may comprise GJEA, GJP or GJPA in a solvent such as but not limited to water, saline, and/or a physiologic buffer, for example, but not limited to, a phosphate buffer, an acetate buffer, a carbonate buffer, a glutamate buffer, a glycinate buffer, a histidine buffer, a lactate buffer, a succinate buffer, a maleate buffer, a tartrate buffer, a Tris buffer, or a citrate buffer.
  • a solvent such as but not limited to water, saline, and/or a physiologic buffer, for example, but not limited to, a phosphate buffer, an acetate buffer, a carbonate buffer, a glutamate buffer, a glycinate buffer, a histidine buffer, a lactate buffer, a succinate buffer, a maleate buffer, a tartrate buffer, a Tris buffer, or a citrate buffer.
  • Non-limiting examples of other ingredients which may optionally be included in pharmaceutical compositions of the invention include albumin, ascorbic acid, sodium bisulphite, sodium metabisulphite, sodium sulphite, thioglycerolm thioglycolic acid, cysteine, ethylene diametetraacetic acid, citric acid/sodium citrate, ethylene glycol, glycerol, glucose, dextran, and/or a surfactant, for example sodium dodecyl sulfate, Polysorbate 80, and/or Polysorbate 20.
  • one or more GJEA, GJP or GJAP may be comprised in an infant nutritional formula, considered a pharmaceutical formulation and also a so-called “nutriceutical” formulation, which may be administered to an infant suffering from NEC or at risk of developing NEC to treat or reduced the risk of NEC in the infant.
  • infant nutritional formula may, for example and not by way of limitation, further comprise one or more of casein, whey protein, soy lecithin, lactose, dextrose, sodium chloride, potassium chloride, calcium carbonate, ferrous sulfate, ascorbic acid, vitamin A, vitamin B6, vitamin B12, vitamin D3, thiamine, vitamin E, and/or vitamin K.
  • the present invention provides for a method of treating NEC in a subject in need of such treatment comprising administering to the subject an effective amount of a GJEA, GJP or GJPA.
  • the present invention provides for a method of treating IBD in a subject in need of such treatment comprising administering to the subject an effective amount of a GJEA, GJP or GJPA.
  • the present invention provides for a method of reducing the risk of occurrence of NEC in a subject in need of such treatment comprising administering to the subject an effective amount of a GJEA, GJP or GJPA.
  • the present invention provides for a method of reducing the risk of occurrence of IBD in a subject in need of such treatment comprising administering to the subject an effective amount of a GJEA, GJP or GJPA.
  • a subject may be a human or non-human subject, including but not limited to a dog, cat, rodent, cow, sheep, goat, horse, or non-human primate.
  • the subject is a human infant.
  • the subject is a human infant born after less than 40 weeks or less than 37 weeks or less than 30 weeks or less than 25 weeks gestation.
  • IBD refers to disorders including but not limited to Crohn's disease, ulcerative colitis, lymphocytic colitis, ischemic colitis, Behçet's disease, diversion colitis, and irritable bowel syndrome.
  • a GJEA, GJP and/or GJPA for example as comprised in a pharmaceutical formulation, may be administered by any route known in the art, including but not limited to oral, intravenous, intraperitoneal, inhalation (nasal and pulmonary), subcutaneous, intramuscular, or rectal. It may be desirable to promote local rather than systemic delivery of a GJEA, GJP or GJPA so as to limit the effect of the agent on gap junctions outside the intestine, for example by selecting a method of administration such as oral or rectal and/or by providing a sustained release formulation.
  • Non-limiting examples of dosages include an amount that results in a local concentration at the intestinal mucosa between 10 and 200 nMol/L, or between 0.125 and 1 mM, or a dosage from 0.05 to 10 mg/kg or between 0.05 and 5 mg/kg or between 0.1 and 1 mg/kg. Dosages may, in non-limiting embodiments, be administered once, twice, three or four times daily, or every other day, or once a week, or once every two weeks, or once a month.
  • Treating means reducing the objective and/or subjective symptoms or signs of the disease being treated, including but not limited to one or more of: a decrease in intestinal tissue viability, malabsorption, diarrhea, bleeding, loss of electrolytes, return to normal activities of daily living and pain. In non-limiting embodiments the reduction is of a magnitude of at least about 20 percent or at least about 30 percent or at least about 40 percent or at least about 50 percent.
  • reducing the risk” of occurrence means a reduction in risk of at least about 10 percent or at least about 20 percent or at least about 30 percent or at least about 40 percent or at least about 50 percent.
  • Enterocyte-specific Cx43 knockout mice were generated using the Cre/loxP system, where Cx43 loxp/loxp mice were generated and crossed with villin-cre mice. The effectiveness of the “knockout” was documented by RT-PCR ( FIG. 1 ). As further confirmation, intestinal tissue of these mice was stained with detectable antibodies directed toward either Cx43 or actin, and the relative absence of Cx43 was apparent ( FIG. 2B ).
  • Enterocytes from the knockout mice were observed to be morphologically and functionally impaired relative to normal enterocytes. As shown in FIGS. 3B and 3A , respectively, the intestinal villi in the Cx43 knockout animals were substantially shortened compared with control intestine. Moreover, enterocytes from the knockout animals (“ ⁇ IEC”) demonstrated lower migration rates ( FIG. 3C ). As shown in FIG. 4 , Cx43 knockout mice were less able to absorb fat from their diet.
  • Enterocyte-specific Cx43 knockout mice were observed to manifest increased susceptibility to NEC.
  • NEC was induced in wild-type control mice and Cx43 knockout animals using a protocol that combines gavage feeding and hypoxia for 4 days.
  • the intestinal tissue of Cx43 knockout mice showed substantially more dramatic alteration relative to wild type (compare FIGS. 5G and 5C ), with substantially greater induction of inducible nitric oxide synthase (iNOS), a marker of inflammation (compare FIG. 5H with FIG. 5D ).
  • the enterocytes in the NEC-induced Cx43 knockout animals exhibited a slower migration rate ( FIG. 6A ) and inflammatory cytokine levels IL-6, TNF, and IL-1 were all substantially higher ( FIG. 6B ).
  • dnCx43 dominant negative mutant Cx43
  • Adenoviruses carrying dnCx43 with detectable marker GFP were constructed, as were adenoviruses carrying only GFP to serve as controls.
  • Virus was introduced into wild-type mice by rectal administration to produce GFP and GFP/dnCx43 animals.
  • Photomicrographs in FIGS. 8B and 8C show enterocytes of mice infected with adenovirus carrying GFP only or GFP and dnCx43, respectively, where the presence of fluorescence in these figures relative to FIG. 8A demonstrates successful viral infection.
  • IEC-6 enterocytes were treated with various concentrations of phorbol myristate acetate (“PMA”) for one hour and then evaluated for the extent of enterocyte gap junction communication by confocal based dye transfer. As shown in FIG. 10A , as concentrations of PMA increased, so did the extent of gap junction communication. Further, IEC-6 cells were treated with the concentration of PMA indicated and assessed for their ability to migrate into a scraped wound over 20 hours. As the concentration of PMA increased, the speed (rate) of enterocyte migration also increased, consistent with an association between gap junction enhancement and migration (and consequent healing) within the intestinal mucosa ( FIG. 10B ).
  • PMA phorbol myristate acetate

Abstract

The present invention relates to methods of reducing the risk of occurrence of, and/or treating, necrotizing enterocolitis (“NEC”) or inflammatory bowel disease (“IBD”) comprising administering, to a subject in need of such treatment, an effective amount of a gap junction enhancing agent (“GJEA”), for example a peptide (“GJP”) or peptide analog (“GJPA”). It is based, at least in part, on the discovery that greater functionality of gap junctions between enterocytes increases their rate of migration and reduces the severity of intestinal inflammation.

Description

PRIORITY
This application is a continuation of International Application No. PCT/US2011/066861, filed Dec. 22, 2011, and claims priority to U.S. Provisional Application No. 61/426,162, filed Dec. 22, 2010, the contents of which are expressly incorporated by reference herein.
SEQUENCE LISTING
The specification further incorporates by reference the Sequence Listing submitted herewith via EFS on Sep. 5, 2013. Pursuant to 37 C.F.R. § 1.52(e)(5), the Sequence Listing text file, identified as 0723960524_SL.txt, is 9,249 bytes and was created on Jul. 29, 2013. The Sequence Listing, electronically filed herewith, does not extend beyond the scope of the specification and thus does not contain new matter.
1. INTRODUCTION
This application relates to agents, particularly peptides and peptide analogs, that enhance the functionality of gap junctions between intestinal enterocytes, and which may be used to treat disorders associated with impaired interenterocyte gap junctions such as necrotizing enterocolitis and inflammatory bowel diseases. These gap junction enhancing peptides may also be used to reduce the risk of occurrence of these disorders.
2. BACKGROUND OF THE INVENTION
Necrotizing enterocolitis (NEC) is a leading cause of death and disability in premature infants. Patients that develop NEC do so suddenly and without warning, and upon surgical exploration of the abdomen, frequently demonstrate large regions of the intestine that are either dead or dying. In over half the cases, patients that develop NEC do not survive their disease, and in survivors, an additional third will develop long term complications related to the initial development of the disease. Currently, there is no specific therapy for NEC, and treatment involves the administration of broad spectrum antibiotics and surgical removal of the dead or dying intestine. Clearly, novel therapeutic approaches to this devastating disease are urgently needed.
In seeking to understand the pathogenesis of NEC, as well as to define novel therapeutic approaches for this disease, it was found that NEC is characterized by impaired enterocyte migration along the crypt-villus axis. The impaired enterocyte migration results in persistent mucosal defects, bacterial translocation, and the development of systemic sepsis which leads to death in many cases. In seeking to define the mechanisms that regulate enterocyte migration in both mice and humans, it was found that enterocytes migrate together as sheets, and that enterocyte migration is dependent upon intact inter-enterocyte communication via Cx43-mediated gap junctions (1, 2). Furthermore, the release of the pro-inflammatory cytokine interferon gamma (IFNγ) plays a critical role in the impairment of mucosal healing in part by inhibiting gap junctions between enterocytes (1, 2).
In other studies, it has been shown that human inflammatory bowel disease (“IBD”) is associated with impaired gap junctions within the intestinal mucosa (1, 2). Enterocyte migration is impaired in IBD just as it is in NEC.
Gap junctions are intercellular channels that exist between adjacent cells which allow the transfer of small molecules between adjoining cells. Each gap junction channel is comprised of a pair of hexameric arrays of individual subunits called connexins, of which the most widely expressed isoform is connexin-43 (Cx43; 2, 3, 4). The function of gap junctions is regulated in part through phosphorylation of the individual connexin molecules, which serves to regulate the localization of the channels at the plasma membrane as well as to regulate the channel through gating (5, 6).
3. SUMMARY OF THE INVENTION
The present invention relates to methods of reducing the risk of occurrence of, and/or treating, necrotizing enterocolitis (“NEC”) or inflammatory bowel disease (“IBD”) comprising administering, to a subject in need of such treatment, an effective amount of a gap junction enhancing agent (“GJEA”), for example a peptide (“GJP”) or peptide analog (“GRA”). It is based, at least in part, on the discovery that greater functionality of gap junctions between enterocytes increases their rate of migration and reduces the severity of intestinal inflammation.
4. BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1. Cx43 knockout “cko” mice were generated. RT-PCR demonstrated the absence of Cx43.
FIG. 2A-B. Immunohistochemistry studies using fluorescently labeled antibodies directed toward Cx43 and actin demonstrate (A) the presence of both proteins in wild-type intestinal villi and (B) the absence of Cx43 in the villi of knock-out animals having the selective deletion of Cx43.
FIG. 3A-C. (A) Photomicrograph of intestinal villi in a wild-type mouse. (B) Photomicrograph of intestinal villi in a Cx43 knockout mouse. (C) migration rate of enterocytes in wild-type versus Cx43 knockout (“ΔIEC”).
FIG. 4. Absorption of fat, protein and glucose by WT versus Cx43 knockout mouse intestine.
FIG. 5A-H. (A) Photomicrograph of intestinal tissue of a healthy wild-type mouse; (B) photomicrograph of intestinal tissue of a healthy wild-type mouse stained to show expression of inducible nitric oxide synthase (“iNOS”); (C) photomicrograph of intestinal tissue of a wild-type mouse having NEC; (D) photomicrograph of intestinal tissue of a wild-type mouse having NEC stained to show expression of iNOS; (E) photomicrograph of intestinal tissue of a Cx43 knockout mouse; (F) photomicrograph of intestinal tissue of a Cx43 knockout mouse stained to show expression of iNOS; (G) photomicrograph of intestinal tissue of a Cx43 knockout mouse having NEC; and (H) photomicrograph of intestinal tissue of a Cx43 knockout mouse with NEC stained to show expression of iNOS.
FIG. 6A-B. (A) Migration rate of enterocytes from either wild-type (“WT”) healthy (control) mice, wild-type mice having NEC, Cx43 knockout mice (“ΔIEC”), or Cx43 mice having NEC. (B) Cytokine levels of either wild-type or Cx43 knockout mice (“Cx43ΔIEC”), having NEC.
FIG. 7A-B. Cx43 expression in (A) premature (gestation day 15.5) or (B) postnatal (day 10) murine intestinal cells, as shown using a fluorescent anti-Cx43 antibody.
FIG. 8A-E. (A) Photomicrograph of control murine intestine. (B) Photomicrograph of murine intestine after exposure to adenoviral vector carrying Green Fluorescent Protein (“GFP”) gene. (C) Photomicrograph of murine intestine after exposure to adenoviral vector carrying GFP and dominant negative Cx43 mutant protein (“dnCx43”). (D) Expression of GFP in murine intestine in the presence (“DN”) or absence (“WT”) of dnCx43. (E) Colitis severity in murine intestine in the presence (“DN”) or absence (“WT”) of dnCx43.
FIG. 9A-C. Surface mucosa of murine intestine, either (A) control; (B) after exposure to adenoviral vector carrying GFP; or (C) after exposure to adenoviral vector carrying GFP and dnCx43.
FIG. 10A-B. (A) IEC-6 enterocytes were treated with the concentrations of phorbol myristate acetate (“PMA”) indicated for one hour and then evaluated for the extent of enterocyte gap junction communication by confocal based dye transfer. The untreated cells represent the 100% dye transfer. *p<0.05 versus untreated cells. (B) IEC-6 cells were treated with the concentration of PMA indicated and assessed for their ability to migrate into a scraped wound over 20 hours. *p<0.05 vs. untreated cells. Representative of two separate experiments.
5. DETAILED DESCRIPTION OF THE INVENTION
For clarity of description, and not by way of limitation, the detailed description of the invention is divided into the following subsections:
(i) gap junction enhancing agents;
(ii) assay methods;
(iii) pharmaceutical compositions; and
(iv) methods of treatment.
5.1 Gap Junction Enhancing Agents
Gap junction enhancing agents (“GJEAs”) include gap junction enhancing peptides (“GJPs”) and peptide analogs (“GJPAs”) as well as other compounds such as pharmacologic agents.
Non-limiting examples of pharmacologic agents that are GJEAs include phorbol myristate acetate (“PMA”) and quinoline derivatives as described in United States Patent Publication No. 20090143425, such as, but not limited to, primaquine, mefloquine, PQ2, PQ3, PQ4, PQ5, PQ6, PQ7, PQ8 and/or the compound PQ1:
Figure US10172848-20190108-C00001
Gap Junction enhancing peptides (“GJPs”) that may be used according to the invention include but are not limited to peptides listed in United States Patent Application Publication No. 20090075291 by Delmar et al., which is incorporated by reference in its entirety herein, including: DVPGRDPGYIKGGGSAHARVPFYSHSLNRNRKPSLYQ (SEQ ID NO:1); EIQPRSPLMFSGGGSAHARVPFYSHSAKEARWPRAHR (SEQ ID NO:2); GIAAREPNSHDGGGSAHARVPFYSHSRDLWRKPAKSL (SEQ ID NO:3); WEEPRRPFTMSGGGSAETHARVPFYSHSPMRHRLPGVHL (SEQ ID NO:4); SDDLRSPQLHNGGGSAVPFYSHSHMVRRKPRNPR (SEQ ID NO:5); GHLHLRVPTLKM (SEQ ID NO:6); EFIRSPHSVDWL (SEQ ID NO:7); SQSRNPPMPPPR (SEQ ID NO:8); RRPPYRVPPKLF (SEQ ID NO:9); SLYERHPASTYP (SEQ ID NO:10); HTVSRRPLPSSG (SEQ ID NO:11); RHTHGNLLRFPP (SEQ ID NO:12); RNNLNQTYPERR (SEQ ID NO:13); YSLLPVRPVALT (SEQ ID NO:14); RKPTQSLPTRLV (SEQ ID NO:15); TRRPHKMRSDPL (SEQ ID NO:16); TLTWHTKTPVRP (SEQ ID NO:17); SRQFLHSLDRLP (SEQ ID NO:18); HLHHHLDHRPHR (SEQ ID NO:19); QTPYQARLPAVA (SEQ ID NO:20); WHPHRHHHLQWD (SEQ ID NO:21); RRKPRRKP (SEQ ID NO:22); RNPRNP (SEQ ID NO:23); RRKP (SEQ ID NO:24); RRNP (SEQ ID NO:25); RNP or SDDLRSPQLHNHMVRRKPRNPR (SEQ ID NO:26), and also include other peptides and peptide derivatives that enhance gap junction communication between enterocytes.
One non-limiting example of a GJPA is Compound ZP123 (Rotigaptide (2R, 4S)-1-[(2R)-1-[(2R)-2-acetamido-3-(4-hydroxyphenyl)propanoyl]pyrrolidine-2-carbonyl]-N-[2-[[(2R)-1-[(2-amino-2-oxoethyl)amino]-1-oxopropan-2-yl]amino]-2-oxoethyl]-4-hydroxypyrrolidine-2-carboxamide), having the following formula:
Figure US10172848-20190108-C00002

which enhances gap junction connectivity through increased activation of Cx43. The prototypical compound, Rotigaptide, is stable, has a long half-life, and is in clinical trials as an anti-arrhythmic agent in adults, based upon its documented ability to enhance gap junction connectivity.
Further non-limiting examples of GJPs include pharmacophores of Cx43 based upon the “RXP” series of Cx43-binding peptides. These pharmacophores bind to the carboxyl terminal of Cx43, and share a 34-aa peptide (RXP-E (SEQ ID NO:27)) sequence. Two representatives of this family are a cyclized heptapeptide (called CyRP-71) and a linear octapeptide of sequence RRNYRRNY (SEQ ID NO:28).
Another non-limiting example of a GJP that may be used according to the present invention is Gly-Ala-Gly-Hyp-Pro-Tyr-NH2 (“AAP10”; SEQ ID NO:29) and related sequences such as but not limited to Gly-Pro-Hyp-Gly-Ala-Gly (SEQ ID NO:30) or cyclo[CF(3)C(OH)-Gly-Ala-Gly-Hyp-Pro-Tyr (SEQ ID NO:31).
The present invention further provides for peptides that are between 4 and 100, or between 4 and 50, or between 6 and 100, or between 6 and 50, or between 8 and 100, or between 8 and 50, or between 10 and 100, or between 10 and 50, or between 15 and 35, amino acids long and comprise one or more peptide having SEQ ID NO:1-31.
The present invention further provides for peptides that are between 4 and 100, or between 4 and 50, or between 6 and 100, or between 6 and 50, or between 8 and 100, or between 8 and 50, or between 10 and 100, or between 10 and 50, or between 15 and 35, amino acids long and comprise one or more peptide having a sequence which differs from any of SEQ ID NO:1-31 by no more than one amino acid and has a gap junction enhancing activity that is at least 80 percent of a peptide lacking the difference in sequence.
The present invention further provides for peptide analogs (“GJPA”) such as compound GAP-134 (i.e. (2S,4R)-1-(2-aminoacetyl)-4-benzamido-pyrrolidine-2-carboxylic acid) or other peptide analogs that enhance gap junction communication between enterocytes. Without being bound by any theory, GAP-134 acts by enhancing gap junction conductance without effects on other ion channels, without any apparent changes in transcription or distribution of Cx43. This compound has a shorter half-life than ZP123, which may decrease its effectiveness, but also may limit potential toxicity;
5.2 Assay Methods
The ability of a GJEA, GJP or GJPA to enhance gap junction communication between enterocytes may be determined by any method known in the art, including in vitro and in vivo studies.
As a non-limiting example, gap junction intercellular communication (GJIC) studies, which assess the ability of a GJEA, GJP or GJPA to enhance gap junction communication between enterocytes, may be performed in vitro in cultured enterocytes, using live cell confocal microscopy and fluorescence recovery after photobleaching (FRAP), as in (1, 2). For example, cells may be plated to confluence, and treated with a GJEA, GJP or GJPA for times between 1-4 hours, and the degree of gap junction intercellular communication may be determined using confocal based fluorescence recovery after photobleaching (FRAP), which measures the movement of a fluorescent tracer through gap junctions into an area that has been previously photobleached using a laser, such that the rate and extent to which the photobleached cells fill with the fluorescent dye (termed the “fluorescence recovery”) may provide a direct measure of gap junction activity.
A second non-limiting example of a method for assessing the ability of a GJEA, GJP or GJPA to enhance gap junction communication between enterocytes is single cell microinjection (2), which allows the detection of the extent to which a detectable tracer, for example the 0.4 kilodalton fluorescent gap junction tracer Lucifer yellow, passes from an injected cell to adjacent cells through gap junctions.
As one specific non-limiting example, IEC-6 cells may be used for such studies, as they represent a well validated culture model of the intestine. Non-limiting examples of other cell lines that may be used include HT-29 and CaCO-2 cells.
In certain non-limiting embodiments of the invention, the ability of a GJEA, GJP or GJPA to treat NEC may be assessed in vivo by the following study in mice.
    • a. Power analysis: Sample size estimates may be conducted using the nQuery Advisor 3.0 software (Statistical Solutions, Saugus, Mass.), with α=0.05, and differences in proportions between Groups averaging 0.22 with a variance of 0.029 and an effect size of 0.167 yielding 15 animals per group to attain 80% power. An estimated 20% of the animals would not be expected to survive, therefore the sample size should be increased to 18 to account for mortality loss. Experiments performed in triplicate with 18 mice per group, 4 groups per experiment=216 mice.
    • b. Induction of experimental NEC: NEC may be induced in 10-day old wild-type mice. Mice may be administered 15 g Similac 60/40 (Ross Pediatrics) in 75 mL of Esbilac canine milk replacer (Pet-Ag Inc) as well as hypoxia (5% oxygen for 2 minute prior to each feeding) twice daily for four days. Animals may be fed 200 microliters per 5 grams of mouse body weight by gavage over 2-3 minutes, using a 24-French angio-catheter which is placed into the mouse esophagus under direct vision. Samples of the terminal ileum 2 cm away from the ileocecal valve may then be harvested at day four for analysis. Control (i.e. non NEC) animals may remain with their mothers and receive breast milk.
    • c. Timing of administration of gap junction peptides: animals may be treated with GJEA, GJP or GJPA prior to the induction of experimental NEC; for example at varying doses, twice daily for either 1, 2, 3 or 4 days prior to the induction of NEC. Morbidity in animals that receive GJEA, GJP or GJPA alone should be assessed. Peptides may be administered via the i.p (intraperitoneal) or the p.o. (oral) route
    • d. Assessment of the severity of experimental NEC: The extent of NEC that develops may be assessed by measuring 1) histopathological evidence of mucosal damage. 2) serum IL-6 as determined by ELISA. 3) RT-PCR to assess the expression of IL-6, IL-8 and TNF-α in the mucosal scrapings, for example using mouse specific primers.
In certain non-limiting embodiments of the invention, the ability of a GJEA, GJP or GJPA to treat IBD may be assessed in vivo by the following study in mice.
    • a. Power analysis: Sample size estimates may be conducted using the nQuery Advisor 3.0 software (Statistical Solutions, Saugus, Mass.), with α=0.05, and differences in proportions between Groups averaging 0.22 with a variance of 0.029 and an effect size of 0.167 yielding 15 animals per group to attain 80% power. There is essentially no mortality in the colitis model, and the sample size may be 25 per group. Total mice: experiments in triplicate, 4 groups, 25 per group—300 mice.
    • b. Induction of experimental colitis: Colitis may be induced in 4 week old mice using the dextran sodium sulfate model. 4% DSS may be administered in the drinking water for 5 days, at which point 100% of mice develop inflammation of the mucosa of the colon, associated with infiltration of inflammatory cells.
    • c. Timing of administration of gap junction peptides: animals may be treated with GJEA, GJP or GJPA prior to the induction of colitis; animals may be administered GJEA, GJP or GJPA for varying doses, twice daily for either 1, 2, 3 or 4 days prior to the induction of colitis. Morbidity in animals that receive GJEA, GJP or GJPA alone should be assessed. GJEA, GJP or GJPA may be administered via the i.p or the p.o. route
    • d. Assessment of the severity of experimental colitis: The extent of colitis that develops may be assessed by measuring 1) histopathological evidence of mucosal damage. 2) serum IL-6 as determined by ELISA. 3) RT-PCR to assess the expression of IL-6, IL-8 and TNF-α in the mucosal scrapings, for example using mouse specific primers.
The foregoing methods may be used to determine whether a particular concentration of a GJEA, GJP or GJPA is able to enhance gap junction communication between enterocytes, to reduce the incidence of NEC, to treat NEC, to reduce the incidence of IBD, or to treat IBD.
In non-limiting embodiments of the invention, aGJEA, GJP or GJPA enhances (increases the rate or amount of) communication between adjacent enterocytes by at least about 10 percent, or at least about 20 percent, or at least about 30 percent, or at least about 40 percent, or at least about 50 percent, or at least about 60 percent, or at least about 70 percent, or at least about 80 percent, or at least about 100 percent, relative to the amount of communication under control conditions (e.g. the absence of the GJEA, GJP or GJPA).
5.3 Pharmaceutical Compositions
The present invention provides for a pharmaceutical composition comprising one or more GJEA, GJP or GJPA in a pharmaceutically suitable carrier.
Said pharmaceutical composition may be in solid or liquid form.
In non-limiting embodiments of the invention, a pharmaceutical composition may comprise GJEA, GJP or GJPA which is optionally lyophilized or comprised in micelles or microspheres.
In non-limiting embodiments of the invention, a pharmaceutical composition may comprise GJEA, GJP or GJPA in a solvent such as but not limited to water, saline, and/or a physiologic buffer, for example, but not limited to, a phosphate buffer, an acetate buffer, a carbonate buffer, a glutamate buffer, a glycinate buffer, a histidine buffer, a lactate buffer, a succinate buffer, a maleate buffer, a tartrate buffer, a Tris buffer, or a citrate buffer.
Non-limiting examples of other ingredients which may optionally be included in pharmaceutical compositions of the invention include albumin, ascorbic acid, sodium bisulphite, sodium metabisulphite, sodium sulphite, thioglycerolm thioglycolic acid, cysteine, ethylene diametetraacetic acid, citric acid/sodium citrate, ethylene glycol, glycerol, glucose, dextran, and/or a surfactant, for example sodium dodecyl sulfate, Polysorbate 80, and/or Polysorbate 20.
In a set of specific non-limiting embodiments of the invention, one or more GJEA, GJP or GJAP may be comprised in an infant nutritional formula, considered a pharmaceutical formulation and also a so-called “nutriceutical” formulation, which may be administered to an infant suffering from NEC or at risk of developing NEC to treat or reduced the risk of NEC in the infant. Such infant nutritional formula may, for example and not by way of limitation, further comprise one or more of casein, whey protein, soy lecithin, lactose, dextrose, sodium chloride, potassium chloride, calcium carbonate, ferrous sulfate, ascorbic acid, vitamin A, vitamin B6, vitamin B12, vitamin D3, thiamine, vitamin E, and/or vitamin K.
5.4 Methods of Treatment
In non-limiting embodiments, the present invention provides for a method of treating NEC in a subject in need of such treatment comprising administering to the subject an effective amount of a GJEA, GJP or GJPA.
In other non-limiting embodiments, the present invention provides for a method of treating IBD in a subject in need of such treatment comprising administering to the subject an effective amount of a GJEA, GJP or GJPA.
In other non-limiting embodiments, the present invention provides for a method of reducing the risk of occurrence of NEC in a subject in need of such treatment comprising administering to the subject an effective amount of a GJEA, GJP or GJPA.
In other non-limiting embodiments, the present invention provides for a method of reducing the risk of occurrence of IBD in a subject in need of such treatment comprising administering to the subject an effective amount of a GJEA, GJP or GJPA.
A subject may be a human or non-human subject, including but not limited to a dog, cat, rodent, cow, sheep, goat, horse, or non-human primate. In non-limiting embodiments the subject is a human infant. In non-limiting embodiments the subject is a human infant born after less than 40 weeks or less than 37 weeks or less than 30 weeks or less than 25 weeks gestation.
IBD as that term is used herein refers to disorders including but not limited to Crohn's disease, ulcerative colitis, lymphocytic colitis, ischemic colitis, Behçet's disease, diversion colitis, and irritable bowel syndrome.
A GJEA, GJP and/or GJPA, for example as comprised in a pharmaceutical formulation, may be administered by any route known in the art, including but not limited to oral, intravenous, intraperitoneal, inhalation (nasal and pulmonary), subcutaneous, intramuscular, or rectal. It may be desirable to promote local rather than systemic delivery of a GJEA, GJP or GJPA so as to limit the effect of the agent on gap junctions outside the intestine, for example by selecting a method of administration such as oral or rectal and/or by providing a sustained release formulation.
Non-limiting examples of dosages include an amount that results in a local concentration at the intestinal mucosa between 10 and 200 nMol/L, or between 0.125 and 1 mM, or a dosage from 0.05 to 10 mg/kg or between 0.05 and 5 mg/kg or between 0.1 and 1 mg/kg. Dosages may, in non-limiting embodiments, be administered once, twice, three or four times daily, or every other day, or once a week, or once every two weeks, or once a month.
“Treating” means reducing the objective and/or subjective symptoms or signs of the disease being treated, including but not limited to one or more of: a decrease in intestinal tissue viability, malabsorption, diarrhea, bleeding, loss of electrolytes, return to normal activities of daily living and pain. In non-limiting embodiments the reduction is of a magnitude of at least about 20 percent or at least about 30 percent or at least about 40 percent or at least about 50 percent.
In non-limiting embodiments, “reducing the risk” of occurrence means a reduction in risk of at least about 10 percent or at least about 20 percent or at least about 30 percent or at least about 40 percent or at least about 50 percent.
6. EXAMPLE 1: SUSCEPTIBILITY TO NEC OF CX43 KNOCKOUT MICE
Enterocyte-specific Cx43 knockout mice were generated using the Cre/loxP system, where Cx43 loxp/loxp mice were generated and crossed with villin-cre mice. The effectiveness of the “knockout” was documented by RT-PCR (FIG. 1). As further confirmation, intestinal tissue of these mice was stained with detectable antibodies directed toward either Cx43 or actin, and the relative absence of Cx43 was apparent (FIG. 2B).
Enterocytes from the knockout mice were observed to be morphologically and functionally impaired relative to normal enterocytes. As shown in FIGS. 3B and 3A, respectively, the intestinal villi in the Cx43 knockout animals were substantially shortened compared with control intestine. Moreover, enterocytes from the knockout animals (“ΔIEC”) demonstrated lower migration rates (FIG. 3C). As shown in FIG. 4, Cx43 knockout mice were less able to absorb fat from their diet.
Enterocyte-specific Cx43 knockout mice were observed to manifest increased susceptibility to NEC. NEC was induced in wild-type control mice and Cx43 knockout animals using a protocol that combines gavage feeding and hypoxia for 4 days. When exposed to the same conditions, the intestinal tissue of Cx43 knockout mice showed substantially more dramatic alteration relative to wild type (compare FIGS. 5G and 5C), with substantially greater induction of inducible nitric oxide synthase (iNOS), a marker of inflammation (compare FIG. 5H with FIG. 5D). The enterocytes in the NEC-induced Cx43 knockout animals exhibited a slower migration rate (FIG. 6A) and inflammatory cytokine levels IL-6, TNF, and IL-1 were all substantially higher (FIG. 6B).
The above findings support an association between NEC and Cx43 deficiency and gap junction dysfunction.
7. EXAMPLE 2: CX43 IN PREMATURE BOWEL
It was observed that Cx43 expression is reduced in the eneterocytes of mouse embryos on day e15.5 (FIG. 7A) relative to eneterocytes of mice at postnatal day 10 (FIG. 7B). As the incidence of NEC is increased in premature, versus full term, human infants, the observation of lower levels of Cx43 in the “premature” intestine is consistent with a model in which Cx43 deficiency and consequent gap junction dysfunction increases susceptibility to NEC.
8. EXAMPLE 3: HIGHER LEVELS OF CX43 CORRELATE WITH LESS COLITIS
Experiments were performed using adenovirus to introduce dominant negative mutant Cx43 (“dnCx43”) into murine enterocytes and thereby create a functional Cx43 deficiency. Adenoviruses carrying dnCx43 with detectable marker GFP were constructed, as were adenoviruses carrying only GFP to serve as controls. Virus was introduced into wild-type mice by rectal administration to produce GFP and GFP/dnCx43 animals. Photomicrographs in FIGS. 8B and 8C show enterocytes of mice infected with adenovirus carrying GFP only or GFP and dnCx43, respectively, where the presence of fluorescence in these figures relative to FIG. 8A demonstrates successful viral infection.
Next, colitis was induced in GFP, GFP/dnCx43 and control animals by administration of dextran sulphate sodium. Cx43 deficiency and consequent gap junction dysfunction and susceptibility to NEC. Visualization of the intestinal mucosa in these animals is shown in FIG. 9A-C. The most severe colitis was observed GFP/dn-Cx43 mice (FIGS. 8E and 9C).
According to these studies, the relatively greater amount of Cx43 in animals that had not received dnCx43 was protective against colitis.
9. EXAMPLE 4: GAP JUNCTION ENHANCER PMA PROMOTES ENTEROCYTE MIGRATION
IEC-6 enterocytes were treated with various concentrations of phorbol myristate acetate (“PMA”) for one hour and then evaluated for the extent of enterocyte gap junction communication by confocal based dye transfer. As shown in FIG. 10A, as concentrations of PMA increased, so did the extent of gap junction communication. Further, IEC-6 cells were treated with the concentration of PMA indicated and assessed for their ability to migrate into a scraped wound over 20 hours. As the concentration of PMA increased, the speed (rate) of enterocyte migration also increased, consistent with an association between gap junction enhancement and migration (and consequent healing) within the intestinal mucosa (FIG. 10B).
7. REFERENCES
  • 1. Leaphart et al., 2008, Interferon-gamma inhibits enterocyte migration by reversibly displacing connexin43 from lipid rafts. Am J Physiol Gastrointest Liver Physiol 295: G559-569.
  • 2. Leaphart et al., 2007, Interferon-[gamma] inhibits intestinal restitution by preventing gap junction communication between enterocytes. Gastroenterology 132: 2395-2411.
  • 3. Goodenough, 1974, Bulk isolation of mouse hepatocyte gap junctions. Characterization of the principal protein, connexin, J. Cell Biol. 61:557-563.
  • 4. Goodenough, 1975, The structure of cell membranes involved in intervellular communication. Am. J. Clin. Pathol. 63:636-645.
  • 5. Laird, 2005, Connexin phosphorylation as a regulatory event linked to gap junction internalization and degradation. Biochim. Biophys. Acta 1711:172-182.
  • 6. Lampe et al., 2000, Phosphorylation of connexin-43 on serine 368 by protein kinase C regulates gap junction communication. J. Cell Biol. 149:1503-1512.
  • 7. Verma et al., 2009, Novel pharmacophores of connexin43 based on the “RXP” series of Cx43-binding peptides, Circ Res. 105(2):176-84.
Various publications are cited herein, the contents of which are hereby incorporated by reference in their entireties.

Claims (15)

What is claimed is:
1. A method of treating necrotizing enterocolitis in a subject in need of such treatment comprising administering to the subject one or more dose of a gap junction enhancing agent, wherein the one or more dose of the gap junction enhancing agent is between 0.05 mg/kg and 10 mg/kg, and wherein the gap junction enhancing agent is selected from the group consisting of
(a) a peptide comprising a sequence selected from the group consisting of SEQ ID NO:1-31; and
(b) a peptide analog selected from the group consisting of rotigaptide and GAP-134 ((2S,4R)-1-(2-aminoacetyl)-4-benzamido-pyrrolidine-2-carboxylic acid).
2. The method of claim 1 where the gap junction enhancing agent comprises a peptide having a sequence selected from the group consisting of SEQ ID NO:1-31.
3. The method of claim 1 where the gap junction enhancing agent is rotigaptide.
4. A method of reducing the risk of occurrence of necrotizing enterocolitis in a subject in need of such treatment comprising administering to the subject one or more dose of a gap junction enhancing agent, wherein the one or more dose of the gap junction enhancing agent is between 0.05 mg/kg and 10 mg/kg, and wherein the gap junction enhancing agent is selected from the group consisting of
(a) a peptide comprising a sequence selected from the group consisting of SEQ ID NO:1-31; and
(b) a peptide analog selected from the group consisting of rotigaptide and GAP-134 ((2S,4R)-1-(2-aminoacetyl)-4-benzamido-pyrrolidine-2-carboxylic acid).
5. The method of claim 4 where the gap junction enhancing agent comprises a peptide having a sequence selected from the group consisting of SEQ ID NO:1-31.
6. The method of claim 4 where the gap junction enhancing agent is rotigaptide.
7. A method of inducing increased migration of enterocytes in a subject diagnosed with necrotizing enterocolitis, the method comprising administering to the subject one or more dose of a gap junction enhancing agent, wherein the one or more dose of the gap junction enhancing agent is between 0.05 mg/kg and 10 mg/kg, and wherein the gap junction enhancing agent is selected from the group consisting of
(a) a peptide comprising a sequence selected from the group consisting of SEQ ID NO:1-31; and
(b) a peptide analog selected from the group consisting of rotigaptide and GAP-134 ((2S,4R)-1-(2-aminoacetyl)-4-benzamido-pyrrolidine-2-carboxylic acid).
8. The method of claim 7 where the gap junction enhancing agent comprises a peptide having a sequence selected from the group consisting of SEQ ID NO:1-31.
9. The method of claim 7 where the gap junction enhancing agent is rotigaptide.
10. The method of claim 1, wherein the one or more dose of the gap junction enhancing agent is between 0.05 mg/kg and 5 mg/kg.
11. The method of claim 10, wherein the one or more dose of the gap junction enhancing agent is between 0.1 mg/kg and 1 mg/kg.
12. The method of claim 4, wherein the one or more dose of the gap junction enhancing agent is between 0.05 mg/kg and 5 mg/kg.
13. The method of claim 12, wherein the one or more dose of the gap junction enhancing agent is between 0.1 mg/kg and 1 mg/kg.
14. The method of claim 7, wherein the one or more dose of the gap junction enhancing agent is between 0.05 mg/kg and 5 mg/kg.
15. The method of claim 14, wherein the one or more dose of the gap junction enhancing agent is between 0.1 mg/kg and 1 mg/kg.
US13/921,865 2010-12-22 2013-06-19 Gap junction-enhancing agents for treatment of necrotizing enterocolitis and inflammatory bowel disease Active US10172848B2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US13/921,865 US10172848B2 (en) 2010-12-22 2013-06-19 Gap junction-enhancing agents for treatment of necrotizing enterocolitis and inflammatory bowel disease
US16/194,009 US20190070168A1 (en) 2010-12-22 2018-11-16 Gap junction-enhancing agents for treatment of necrotizing enterocolitis and inflammatory bowel disease

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201061426162P 2010-12-22 2010-12-22
PCT/US2011/066861 WO2012088425A2 (en) 2010-12-22 2011-12-22 Gap junction-enhancing agents for treatment of necrotizing enterocolitis and inflammatory bowel disease
US13/921,865 US10172848B2 (en) 2010-12-22 2013-06-19 Gap junction-enhancing agents for treatment of necrotizing enterocolitis and inflammatory bowel disease

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/066861 Continuation WO2012088425A2 (en) 2010-12-22 2011-12-22 Gap junction-enhancing agents for treatment of necrotizing enterocolitis and inflammatory bowel disease

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/194,009 Continuation US20190070168A1 (en) 2010-12-22 2018-11-16 Gap junction-enhancing agents for treatment of necrotizing enterocolitis and inflammatory bowel disease

Publications (2)

Publication Number Publication Date
US20130345154A1 US20130345154A1 (en) 2013-12-26
US10172848B2 true US10172848B2 (en) 2019-01-08

Family

ID=46314941

Family Applications (2)

Application Number Title Priority Date Filing Date
US13/921,865 Active US10172848B2 (en) 2010-12-22 2013-06-19 Gap junction-enhancing agents for treatment of necrotizing enterocolitis and inflammatory bowel disease
US16/194,009 Abandoned US20190070168A1 (en) 2010-12-22 2018-11-16 Gap junction-enhancing agents for treatment of necrotizing enterocolitis and inflammatory bowel disease

Family Applications After (1)

Application Number Title Priority Date Filing Date
US16/194,009 Abandoned US20190070168A1 (en) 2010-12-22 2018-11-16 Gap junction-enhancing agents for treatment of necrotizing enterocolitis and inflammatory bowel disease

Country Status (2)

Country Link
US (2) US10172848B2 (en)
WO (1) WO2012088425A2 (en)

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8518903B2 (en) 2007-04-19 2013-08-27 University of Pittsburgh—of the Commonwealth System of Higher Education Use of toll-like receptor-9 agonists
US9072760B2 (en) 2010-09-24 2015-07-07 University of Pittsburgh—of the Commonwealth System of Higher Education TLR4 inhibitors for the treatment of human infectious and inflammatory disorders
US10668092B2 (en) 2010-09-24 2020-06-02 The John Hopkins University Compositions and methods for treatment of inflammatory disorders
WO2012088425A2 (en) 2010-12-22 2012-06-28 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Gap junction-enhancing agents for treatment of necrotizing enterocolitis and inflammatory bowel disease
WO2014052453A1 (en) 2012-09-25 2014-04-03 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Oral therapy of necrotizing enterocolitis
GB201512139D0 (en) * 2015-07-10 2015-08-19 Zealand Pharma As Methods of treatment
CN110709096B (en) 2017-05-05 2023-10-31 泽兰德制药公司 Cell gap junction communication regulator and application thereof in treatment of diabetic eye disease
EP3930735A1 (en) * 2019-02-26 2022-01-05 The Regents of the University of Colorado, a body corporate Method and composition for treating gastrointestinal inflammatory disorders

Citations (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4357322A (en) 1980-07-29 1982-11-02 Syntex (U.S.A.) Inc. Method of preventing, reducing or inhibiting inflammation
JPH01180894A (en) 1988-01-07 1989-07-18 Wako Pure Chem Ind Ltd Novel production of amino sugar derivative
US5506204A (en) 1991-11-19 1996-04-09 Peptech (Uk) Limited Muramyl compounds for treatment of septic shock
WO1998018810A1 (en) 1996-10-30 1998-05-07 The University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US5756718A (en) 1991-10-11 1998-05-26 Eisai Co., Ltd. Anti-endotoxin compounds
WO1998037919A1 (en) 1997-02-28 1998-09-03 University Of Iowa Research Foundation USE OF NUCLEIC ACIDS CONTAINING UNMETHYLATED CpG DINUCLEOTIDE IN THE TREATMENT OF LPS-ASSOCIATED DISORDERS
WO1998052581A1 (en) 1997-05-20 1998-11-26 Ottawa Civic Hospital Loeb Research Institute Vectors and methods for immunization or therapeutic protocols
WO1999033488A2 (en) 1997-12-24 1999-07-08 Smithkline Beecham Biologicals S.A. Adjuvanted vaccine formulation
US5962636A (en) 1998-08-12 1999-10-05 Amgen Canada Inc. Peptides capable of modulating inflammatory heart disease
WO2000061555A1 (en) 1999-04-12 2000-10-19 Gerhard Eisenbrand Indigoid bisindole derivatives
US6218371B1 (en) 1998-04-03 2001-04-17 University Of Iowa Research Foundation Methods and products for stimulating the immune system using immunotherapeutic oligonucleotides and cytokines
US6239116B1 (en) 1994-07-15 2001-05-29 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US6406705B1 (en) 1997-03-10 2002-06-18 University Of Iowa Research Foundation Use of nucleic acids containing unmethylated CpG dinucleotide as an adjuvant
US6429199B1 (en) 1994-07-15 2002-08-06 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules for activating dendritic cells
US6544518B1 (en) 1999-04-19 2003-04-08 Smithkline Beecham Biologicals S.A. Vaccines
US6558670B1 (en) 1999-04-19 2003-05-06 Smithkline Beechman Biologicals S.A. Vaccine adjuvants
US6613751B2 (en) 2000-02-23 2003-09-02 The Regents Of The University Of California Method for treating inflammatory bowel disease and other forms of gastrointestinal inflammation
US6652392B2 (en) 2000-12-22 2003-11-25 Bridgestone Sports Co., Ltd. Multi-piece solid golf ball
WO2004096156A2 (en) 2003-04-29 2004-11-11 Centocor, Inc. Toll-like receptor 9 effector agents and uses thereof
US20050250726A1 (en) 2001-03-29 2005-11-10 University Of Iowa Research Foundation Immunostimulatory nucleic acid for treatment of non-allergic inflammatory diseases
US7038029B2 (en) 2002-05-30 2006-05-02 Immunotech S.A. Immunostimulatory oligonucleotides and uses thereof
US7049302B1 (en) 1998-08-10 2006-05-23 Antigenics Inc. Compositions of CPG and saponin adjuvants and uses thereof
WO2006092049A1 (en) 2005-03-01 2006-09-08 Simon Fraser University Selective glycosidase inhibitors, methods of making inhibitors, and uses thereof
US20060211752A1 (en) 2004-03-16 2006-09-21 Kohn Leonard D Use of phenylmethimazoles, methimazole derivatives, and tautomeric cyclic thiones for the treatment of autoimmune/inflammatory diseases associated with toll-like receptor overexpression
US20060241040A1 (en) 2005-04-06 2006-10-26 Alberto Visintin Methods of treating disorders associated with toll-like receptor 4 (TLR4) signalling
US7129222B2 (en) 2000-03-10 2006-10-31 Dynavax Technologies Corporation Immunomodulatory formulations and methods for use thereof
US20070004654A1 (en) 2005-02-22 2007-01-04 Eyal Raz Methods of treating gastrointestinal inflammation
US7250397B2 (en) * 2000-02-23 2007-07-31 Zealand Pharma A/S Antiarrhythmic peptides
WO2007106886A2 (en) 2006-03-15 2007-09-20 Trustees Of Tufts College Fluorinated carbohydrates and their use in tumor visualization, tissue engineering, and cancer chemotherapy
WO2007120368A2 (en) 2006-01-09 2007-10-25 The Regents Of The University Of California Immunostimulatory combinations for vaccine adjuvants
US7348316B2 (en) 2000-06-09 2008-03-25 Eisai R&D Management Co., Ltd. Use of an anti-endotoxin drug in the prevention and treatment of disease
WO2008131074A1 (en) 2007-04-19 2008-10-30 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Use of toll-like receptor-9 agonists, toll-like receptor-4 antagonists, and/or nuclear oligomerization domain-2 agonists for the treatment or prevention of toll-like receptor-4-associated disorders
US20090010902A1 (en) 2007-07-06 2009-01-08 Toa Pharmaceutical Co., Ltd. TLR4 Transcription Activity Suppressor
US7744884B2 (en) 2003-12-10 2010-06-29 Novimmune S.A. Neutralizing antibodies and methods of use thereof
US7851451B2 (en) 2004-03-12 2010-12-14 Mti Meta Tech Inc. Formulations for mediating inflammatory bowel disorders
US20120077868A1 (en) 2007-04-19 2012-03-29 Hackam David J Use of Toll-like receptor-9 agonists, Toll-like receptor-4 antagonists, and/or nuclear oligomerization domain-2 agonists for the treatment or prevention of Toll-like receptor-4-associated disorders
US20130281395A1 (en) 2010-09-24 2013-10-24 University of Pittsburgh - Of the Commonwealth System of Higher Education Novel tlr4 inhibitors for the treatment of human infectious and inflammatory disorders
US20130345154A1 (en) 2010-12-22 2013-12-26 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Gap Junction-Enhancing Agents for Treatment of Necrotizing Enterocolitis and Inflammatory Bowel Disease
US20140086982A1 (en) 2012-09-25 2014-03-27 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Oral therapy of necrotizing enterocolitis
US20140377238A1 (en) 2011-06-20 2014-12-25 H.J. Heinz Company Probiotic compositions and methods

Patent Citations (54)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4357322A (en) 1980-07-29 1982-11-02 Syntex (U.S.A.) Inc. Method of preventing, reducing or inhibiting inflammation
JPH01180894A (en) 1988-01-07 1989-07-18 Wako Pure Chem Ind Ltd Novel production of amino sugar derivative
US5756718A (en) 1991-10-11 1998-05-26 Eisai Co., Ltd. Anti-endotoxin compounds
US5506204A (en) 1991-11-19 1996-04-09 Peptech (Uk) Limited Muramyl compounds for treatment of septic shock
US6207646B1 (en) 1994-07-15 2001-03-27 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
US6429199B1 (en) 1994-07-15 2002-08-06 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules for activating dendritic cells
US6239116B1 (en) 1994-07-15 2001-05-29 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
WO1998018810A1 (en) 1996-10-30 1998-05-07 The University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
WO1998037919A1 (en) 1997-02-28 1998-09-03 University Of Iowa Research Foundation USE OF NUCLEIC ACIDS CONTAINING UNMETHYLATED CpG DINUCLEOTIDE IN THE TREATMENT OF LPS-ASSOCIATED DISORDERS
US6214806B1 (en) 1997-02-28 2001-04-10 University Of Iowa Research Foundation Use of nucleic acids containing unmethylated CPC dinucleotide in the treatment of LPS-associated disorders
US6406705B1 (en) 1997-03-10 2002-06-18 University Of Iowa Research Foundation Use of nucleic acids containing unmethylated CpG dinucleotide as an adjuvant
US6339068B1 (en) 1997-05-20 2002-01-15 University Of Iowa Research Foundation Vectors and methods for immunization or therapeutic protocols
WO1998052581A1 (en) 1997-05-20 1998-11-26 Ottawa Civic Hospital Loeb Research Institute Vectors and methods for immunization or therapeutic protocols
WO1999033488A2 (en) 1997-12-24 1999-07-08 Smithkline Beecham Biologicals S.A. Adjuvanted vaccine formulation
US6218371B1 (en) 1998-04-03 2001-04-17 University Of Iowa Research Foundation Methods and products for stimulating the immune system using immunotherapeutic oligonucleotides and cytokines
US20020064515A1 (en) 1998-04-03 2002-05-30 Krieg Arthur M. Methods and products for stimulating the immune system using immunotherapeutic oligonucleotides and cytokines
US7049302B1 (en) 1998-08-10 2006-05-23 Antigenics Inc. Compositions of CPG and saponin adjuvants and uses thereof
US6034230A (en) 1998-08-12 2000-03-07 Amgen Canada Inc. Nucleic acids encoding myocardial peptides
US5962636A (en) 1998-08-12 1999-10-05 Amgen Canada Inc. Peptides capable of modulating inflammatory heart disease
WO2000061555A1 (en) 1999-04-12 2000-10-19 Gerhard Eisenbrand Indigoid bisindole derivatives
US6558670B1 (en) 1999-04-19 2003-05-06 Smithkline Beechman Biologicals S.A. Vaccine adjuvants
US6544518B1 (en) 1999-04-19 2003-04-08 Smithkline Beecham Biologicals S.A. Vaccines
US6613751B2 (en) 2000-02-23 2003-09-02 The Regents Of The University Of California Method for treating inflammatory bowel disease and other forms of gastrointestinal inflammation
US7250397B2 (en) * 2000-02-23 2007-07-31 Zealand Pharma A/S Antiarrhythmic peptides
US7129222B2 (en) 2000-03-10 2006-10-31 Dynavax Technologies Corporation Immunomodulatory formulations and methods for use thereof
US7183111B2 (en) 2000-03-10 2007-02-27 Dynavax Technologies Corporation Immunomodulatory formulations and methods for use thereof
US7348316B2 (en) 2000-06-09 2008-03-25 Eisai R&D Management Co., Ltd. Use of an anti-endotoxin drug in the prevention and treatment of disease
US6652392B2 (en) 2000-12-22 2003-11-25 Bridgestone Sports Co., Ltd. Multi-piece solid golf ball
US20050250726A1 (en) 2001-03-29 2005-11-10 University Of Iowa Research Foundation Immunostimulatory nucleic acid for treatment of non-allergic inflammatory diseases
US7038029B2 (en) 2002-05-30 2006-05-02 Immunotech S.A. Immunostimulatory oligonucleotides and uses thereof
WO2004096156A2 (en) 2003-04-29 2004-11-11 Centocor, Inc. Toll-like receptor 9 effector agents and uses thereof
US7744884B2 (en) 2003-12-10 2010-06-29 Novimmune S.A. Neutralizing antibodies and methods of use thereof
US7851451B2 (en) 2004-03-12 2010-12-14 Mti Meta Tech Inc. Formulations for mediating inflammatory bowel disorders
US20060211752A1 (en) 2004-03-16 2006-09-21 Kohn Leonard D Use of phenylmethimazoles, methimazole derivatives, and tautomeric cyclic thiones for the treatment of autoimmune/inflammatory diseases associated with toll-like receptor overexpression
US20070004654A1 (en) 2005-02-22 2007-01-04 Eyal Raz Methods of treating gastrointestinal inflammation
WO2006092049A1 (en) 2005-03-01 2006-09-08 Simon Fraser University Selective glycosidase inhibitors, methods of making inhibitors, and uses thereof
US20060241040A1 (en) 2005-04-06 2006-10-26 Alberto Visintin Methods of treating disorders associated with toll-like receptor 4 (TLR4) signalling
WO2007120368A2 (en) 2006-01-09 2007-10-25 The Regents Of The University Of California Immunostimulatory combinations for vaccine adjuvants
WO2007106886A2 (en) 2006-03-15 2007-09-20 Trustees Of Tufts College Fluorinated carbohydrates and their use in tumor visualization, tissue engineering, and cancer chemotherapy
US20120077868A1 (en) 2007-04-19 2012-03-29 Hackam David J Use of Toll-like receptor-9 agonists, Toll-like receptor-4 antagonists, and/or nuclear oligomerization domain-2 agonists for the treatment or prevention of Toll-like receptor-4-associated disorders
US8518903B2 (en) 2007-04-19 2013-08-27 University of Pittsburgh—of the Commonwealth System of Higher Education Use of toll-like receptor-9 agonists
US20080311112A1 (en) 2007-04-19 2008-12-18 Hackam David J Use of toll-like receptor-9 agonists, toll-like receptor-4 antagonists, and/or nuclear oligomerization domain-2 agonists for the treatment or prevention of toll-like receptor-4-associated disorders
WO2008131074A1 (en) 2007-04-19 2008-10-30 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Use of toll-like receptor-9 agonists, toll-like receptor-4 antagonists, and/or nuclear oligomerization domain-2 agonists for the treatment or prevention of toll-like receptor-4-associated disorders
US8188058B2 (en) 2007-04-19 2012-05-29 University of Pittsburgh—of the Commonwealth System of Higher Education Use of toll-like receptor-9 agonists, toll-like receptor-4 antagonists, and/or nuclear oligomerization domain-2 agonists for the treatment or prevention of toll-like receptor-4-associated disorders
US20130072547A1 (en) 2007-04-19 2013-03-21 University of Pittsburgh - of the Commonwealth of Higher Education Use of toll-like receptor-9 agonists, toll-like receptor-4 antagonists, and/or nuclear oligomerization domain-2 agonists for the treatment of prevention of toll-like receptor-4-associated disorders
US8518905B2 (en) 2007-04-19 2013-08-27 University Of Pittsburgh-Of The Commonwealth System Of Higher Education Use of toll-like receptor-9 agonists, toll-like receptor-4 antagonists, and/or nuclear oligomerization domain-2 agonists for the treatment of prevention of toll-like receptor-4-associated disorders
US20150056217A1 (en) 2007-04-19 2015-02-26 University of Pittsburgh - of the Commonwealth System of Highter Education Use of toll-like receptor-9 agonists, toll-like receptor-4 antagonists, and/or nuclear oligomerization domain-2 agonists for the treatment or prevention of toll-like receptor-4 associated disorders
US20090010902A1 (en) 2007-07-06 2009-01-08 Toa Pharmaceutical Co., Ltd. TLR4 Transcription Activity Suppressor
US20130281395A1 (en) 2010-09-24 2013-10-24 University of Pittsburgh - Of the Commonwealth System of Higher Education Novel tlr4 inhibitors for the treatment of human infectious and inflammatory disorders
US9072760B2 (en) 2010-09-24 2015-07-07 University of Pittsburgh—of the Commonwealth System of Higher Education TLR4 inhibitors for the treatment of human infectious and inflammatory disorders
US20130345154A1 (en) 2010-12-22 2013-12-26 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Gap Junction-Enhancing Agents for Treatment of Necrotizing Enterocolitis and Inflammatory Bowel Disease
US20140377238A1 (en) 2011-06-20 2014-12-25 H.J. Heinz Company Probiotic compositions and methods
US20140086982A1 (en) 2012-09-25 2014-03-27 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Oral therapy of necrotizing enterocolitis
WO2014052453A1 (en) 2012-09-25 2014-04-03 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Oral therapy of necrotizing enterocolitis

Non-Patent Citations (213)

* Cited by examiner, † Cited by third party
Title
Abreu et al., 2005, "TLR Signaling in the Gut in Health and Disease." J Immunol 174:4453-4460.
Abreu, "Toll-like receptor signalling in the intestinal epithelium: how bacterial recognition shapes intestinal function," Nature Reviews/Immunology, Feb. 2010, 10:131-143.
Achkar, "Ulcerative colitis: Responding to the challenges", Cleveland Clinic J. Med., 2007; 74(9):657-660.
Afrazi et al. "New insights into the pathogenesis and treatment of necrotizing enterocolitis: Toll-like receptors and beyond", Pediatr Res., 2011; 69:183-188.
Afrazi et al., "Intracellular heat shock protein-70 negatively regulates TLR4 signaling in the newborn intestinal epithelium", J. Immunol., 2012, 188:4543-4557.
Aki Tsukioka, "Eisai Successfully Completes Phase II Trial of Eritoran, Drug Candidate for Severe Sepsis." JCN Network, Aug. 30, 2005 p. 1. Downloaded on Nov. 20, 2009 from http://www.japancorp.net/printarticle.asp?Art_ID=10765.
Amer et al., "Platelet-activating factor concentration in the stool of human newborns: effects of enteral feeding and neonatal necrotizing enterocolitis", Biol Neonate, 2004; 85:159-166.
Anand et al., 2007, "The Role of the Intestinal Barrier in the Pathogenesis of Necrotizing Enterocolitis." Shock 27:124-133.
Anderson, 2001, "Infant, neonatal, and postnatal deaths, percent of total deaths, and mortality rates for the 10 leading causes of infant death by race and sex: United States: 1999." National Vital Statistics Reports. 49:73.
Arciero et al., "Modeling the interactions of bacteria and Toll-like receptor-mediated inflammation in necrotizing enterocolitis," Journal of Theoretical Biology, 2013, 321:83-99.
Blakely et al., "Postoperative outcomes of extremely low birth-weight infants with necrotizing enterocolitis or isolated intestinal perforation: a prospective cohort study by the NICHD Neonatal Research Network", Ann Surg. 2005; 241(6):984-989.
Borges et al., "Immune response by nasal delivery of hepatitis B surface and antigen and codelivery of a CpG ODN in alginate coated chitosan nanoparticles", European Journal of Pharmaceutics and Biopharmaceutics, 59:405-416 (2008).
Borzutzky et al., "NOD2-associated diseases: Bridging innate immunity and autoinflammation", Clin Immunol., 2010; 134:251-261.
Caplan et al., "Neonatal necrotizing entercolitis: possible role of probiotic supplementation", Journal of Pediatric Gastroenterology and Nutrition, 30(2):S18-S22 (2000).
Caplan et al., "The platelet activating factor receptor antagonist WEB 2170 prevents neonatal necrotizing enterocolitis in rats", J Pediatr Gastroenterol Nutr. 1997; 24:296-301.
Caplan et al., "The role of recombinant platelet activating factor acetylhydrolase in a neonatal rat model of necrotizing enterocolitis", Pediatr Res., 1997; 42:779-783.
Caradonna et al., "Phagocytosis, killing, lymphocyte-mediated antibacterial activity, serum autoantibodies, and plasma endotoxins in inflammatory bowel disease", Am J Gastroenterol. 2000; 95:1495-1502.
Career Opportunities-Eisai annuonces Phase II results, plans to initiate phase III clinical-Aug. 29, 2005. Downloaded on Apr. 18, 2007 from http://www.eisai.com/view_pressrelease.asp?ID=145&press=124.
Career Opportunities—Eisai annuonces Phase II results, plans to initiate phase III clinical—Aug. 29, 2005. Downloaded on Apr. 18, 2007 from http://www.eisai.com/view_pressrelease.asp?ID=145&press=124.
Cario et al., 2000, "Lipopolysaccharide activates distinct signaling pathways in intestinal epithelial cell lines expressing Toll-like receptors." J Immunol. 164(2):966-72.
Carneiro et al., 2008, "Nod-like proteins in inflammation and disease." J Pathol. 214(2):136-48.
Cavallo et al., 2006 "The expression and function of enterocyte toll like receptor-4 are enhanced by lipopolysaccharide in vitro and during systemic endotoxemia." Association for academic surgery and society of university surgeons-Abstracts. Journal of Surgical Researchvol. 130, Issue 2, p. 232, No. 189.
Cavallo et al., 2006 "The expression and function of enterocyte toll like receptor-4 are enhanced by lipopolysaccharide in vitro and during systemic endotoxemia." Association for academic surgery and society of university surgeons—Abstracts. Journal of Surgical Researchvol. 130, Issue 2, p. 232, No. 189.
Cetin et al., 2004, "Endotoxin inhibits intestinal epithelial restitution through activation of Rho-GTPase and increased focal adhesions." J Biol Chem. 279(23):24592-600. Epub Mar. 30, 2004.
Cetin et al., 2007, "Nitric oxide inhibits enterocyte migration through activation of RhoA-GTPase in a SHP-2-dependent manner." Am J Physiol Gastrointest Liver Physiol 292:G1347-1358.
Chan et al., "Role of LPS/CD14/TLR4-mediated inflammation in necrotizing enterocolitis: pathogenesis and therapeutic implications", World J Gastroenterol., 2009; 15:4745-4752.
Cho et al., 2007, "The genetics of inflammatory bowel disease." Gastroenterology 133:1327-1339.
Creagh et al., 2006, "TLRs, NLRs and RLRs: a trinity of pathogen sensors that co-operate in innate immunity." Trends Immunol. 27(8):352-7. Epub Jun. 27, 2006.
Dai et al., "Extracellular high mobility group box1 (HMGB1) inhibits enterocyte migration via activation of toll like receptor 4 and increased cell-matrix adhesiveness", J Biol Chem., 2010; 285:4995-5002.
Daubenberger, 2007, "TLR9 agonists as adjuvants for prophylactic and therapeutic vaccines." Curr. Opin. Molec. Ther. 9:45-52.
Ding et al., 1998, "Characterization and quantitation of NF-kappaB nuclear translocation induced by interleukin-1 and tumor necrosis factor-alpha. Development and use of a high capacity fluorescence cytometric system." J Biol Chem. 273(44):28897-905.
Diwan et al., "Enhancement of immune responses by co-delivery of a CpG oligodeoxynucleotide and tetanus toxoid in biodegradable nanospheres", J. Control Release, 85(1-3):247-262 (2002).
Duffy et al., "Concordance of bacterial cultures with endotoxin and interleukin-6 in necrotizing enterocolitis", Dig Dis Sci. 1997; 42:359-365.
Ewaschuk et al., 2007, "Surface expression of Toll-like receptor 9 is upregulated on intestinal epithelial cells in response to pathogenic bacterial DNA." Infect Immun. 75(5):2572-9. Epub Feb. 26, 2007.
Ey et al., "Connexin-43 in Intestinal Epithelial Barrier Intercellular Communication through TLR2 Mediates Gap Junctional Injury," J. Biol. Chem. 284:22332-43 (Jun. 15, 2009). *
Ey et al., "TLR2 Mediates Gap Junctional Intercellular Communication through Connexin-43 in Intestinal Epithelial Barrier Injury," J. Biol. Chem. 284:22332-43 (2009). *
Feng et al., "Heparin-binding epidermal growth factor-like growth factor promotes enterocyte migration and proliferation in neonatal rats with necrotizing enterocolitis", J. Pediatr Surg., 2007; 42:214-220.
Feng et al., 2005, "Heparin-binding EGF-like growth factor (HB-EGF) and necrotizing enterocolitis." Semin Pediatr Surg. 14(3):167-74.
Franchi et al., 2008, "Intracellular NOD-like receptors in innate immunity, infection and disease." Cell Microbiol 10:1-8.
Fukata et al., "Cox-2 is regulated by Toll-like receptor-4 (TLR4) signaling: Role in proliferation and apoptosis in the intestine", Gastroenterology, 2006; 131:862-877.
Fukata et al., "Innate immune signaling by Toll-like receptor-4 (TLR4) shapes the inflammatory microenvironment in colitis-associated tumors", Inflamm Bowel Dis. 2009; 15:997-1006.
Fukata et al., "TLR4 signaling in the intestine in health and disease", Biochemical Society Transactions, 35(6):1473-1478 (2007).
Fukata et al., "Toll-like receptor-4 is required for intestinal response to epithelial injury and limiting bacterial translocation in a murine model of acute colitis", Am J Physiol Gastrointest Liver Physiol., 2005; 288:G1055-G1065.
Gagliardi et al., "Necrotising enterocolitis in very low birth weight infants in Italy: incidence and non-nutritional risk factors", J. Pediatr Gastroenterol Nutr., 2008; 47(2):206-210.
Good et al., "Evidence based feeding strategies before and after the development of necrotizing enterocolitis," Expert Rev Clin Immunol., Jul. 2014; 10(7):875-884.
Goodenough, "Bulk isolation of mouse hepatocyte gap junctions. Characterization of the principal protein connexin", J. Cell Biol., 1974; 61: 557-563.
Goodenough, "The structure of cell membranes involved in intercellular communication", Am. J. Clin. Pathol., 1975; 63:636-645.
Grave et al., "New therapies and preventive approaches for necrotizing enterocolitis: report of a research planning workshop", Pediatr Res., 2007; 62:510-514.
Gribar et al., "Reciprocal expression and signaling of TLR4 and TLR9 in the pathogenesis and treatment of necrotizing entercolitis", Journal of Immunologists, 182(1):636-646 (2009).
Gribar et al., 2008, "The role of epithelial Toll-like receptor signaling in the pathogenesis of intestinal inflammation." J Leukoc Biol. 83(3):493-8. Epub Dec. 26, 2007.
Grimm et al., "NOD2 Mutations and Crohn's Disease: Are Paneth Cells and Their Antimicrobial Peptides the Link?" Gut; 53(11): 1558-1560, Nov. 2004, entire document especially p. 2.
Guthrie et al., 2003, "Necrotizing enterocolitis among neonates in the United States." J Perinatol 23:278-285.
Hackam et al., "Mechanisms of gut barrier failure in the pathogenesis of necrotizing enterocolitis: Toll-like receptors throw the switch," Semin Pediatr Surg 22(2): 76-82, May 2013.
Halpern et al., "Reduction of experimental necrotizing enterocolitis with anti-TNF-alpha", Am J Physiol Gastrointest Liver Physiol 290:757-764, 2006, First published Nov. 3, 2005, entire document especially abstract; p. 1.
Halpern et al., 2006, "Reduction of experimental necrotizing enterocolitis with anti-TNF-α." Am J. Physiol Gastrointest Liver Physiol 290, pp. G757-G764.
Henckaerts et al., "NOD/CARD15 Disease Associations Other Than Crohn's Disease," Inflamm Bowel Dis 13(2): 235-241, 2007.
Henry et al., 2005, "Surgical therapy for necrotizing enterocolitis: bringing evidence to the bedside." Semin Pediatr Surg. 14(3):181-90.
Henry et al., 2006, "Laparotomy Versus Peritoneal Drainage for Perforated Necrotizing Enterocolitis." Neoreviews 7:456-462.
Hotta et al., "Lipopolysaccharide-induced colitis in rabbits", Res Exp Med (Berl) 1986; 186:61-69.
Hsueh et al., 2003, "Neonatal necrotizing enterocolitis: clinical considerations and pathogenetic concepts." Pediatr Dev Pathol 6:6-23.
Hugot et al., "Association of NOD2 leucine-rich repeat variants with susceptibility to Crohn's disease." Nature. 411(6837):599-603 (2001).
IEC-6 cells, Sigma Aldrich, accessed Aug. 2, 2016 at URL sigmaaldrich.com/catalog/product/sigma/88071401, 1 page. *
International Search Report for PCT/US2011/053293, dated Apr. 9, 2012.
InvivoGen: Delivering Genes. "TLR9 Ligands." Downloaded on Apr. 16, 2007 from hrrp://www.invivogen.com/family.php?ID=104&ID_cat=2&ID_sscat=9.
Iwasaki et al., "Regulation of adaptive immunity by the innate immune system", Science, 2010; 327:291-295.
Izumi et al., "Platelet-activating factor receptor: gene expression and signal transduction", Biochim Biophys Acta, 1995; 1259:317-333.
Jesse et al., 2006, "Necotrizing enterocolitis: Relationship to Innate Immunity, Clinical Features, and Strategies for Prevention." NeoReviews 7:143-150.
Jilling et al., "The roles of bacteria and TLR4 in rat and murine models of necrotizing enterocolitis." J Immunol. 177(5):3273-82 (2006).
Kanneganti et al., 2007, "Intracellular NOD-like receptors in host defense and disease." Immunity 27:549-559.
Katakura et al., "Toll-like receptor 9-induced type I IFN protects mice from experimental colitis." J Clin Invest. 115(3):695-702. Erratum in: J Clin Invest. 2005 115(4):1100 (2005).
Kitagaki et al., "Oral administration of CpG-ODNs suppresses antigen-induced asthma in mice", British Society for Immunology, Clinical and Experimental Immunology, 143:249-259 (2005).
Knapp, et al., "Thionation: GlcNAc-Thiazoline Triacetate {(3aR,5R,6S,7R,7aR)-5-Acetoxymethyl-6, 7-Diacetoxy-2-Methyl-5,6,7,7a-Tetrahydro-3aH-Pyrano[3,2-d]Thiazole}", Organic Syntheses, 84:68-76 (2007).
Kobayashi et al., "Suppression of murine endotoxin response by E5531, a novel synthetic lipid A antagonist." Antimicrob Agents Chemother. 42(11):2824-9 (1998).
Kosloske, 1994, "Epidemiology of necrotizing enterocolitis." Acta Pediatr. Suppl. 396:2-7.
Krieg, 2006, "Therapeutic potential of Toll-like receptor 9 activation." Nat. Rev. Drug Disc. 5:471-484.
Kruis et al., "Circulating lipid A antibodies despite absence of systemic endotoxemia in patients with Crohn's disease", Dig Dis Sci., 1984; 29:502-507.
Laird, "Connexin phosphorylation as a regulatory event linked to gap junction internalization and degradation", Biochi. Biophys. Acta, 2005; 1711: 172-182.
Lampe et al., "Phosphorylation of connexin-43 on serine 368 by protein kinase C regulates gap junction communication", J. Cell Biol., (2000) 149:1503-1512.
Lavelle et al., "The role of TLRs, NLRs, and RLRs in mucosal innate immunity and homeostasis," Mucosal Immunol 3(1): 17-28, Jan. 2010.
Leapart et al., "Interferon-γ inhibits enterocyte migration by reversibly displacing connexion43 from lipid rafts", Am J Physiol Gastrointest Liver Physiol, 2008; 295:G559-G569.
Leaphart et al., 2007, "Interferon-gamma inhibits intestinal restitution by preventing gap junction communication between enterocytes." Gastroenterology. 132(7):2395-411. Epub Mar. 21, 2007.
Leaphart et al., 2007. "A Critical Role for TLR4 in the Pathogenesis of Necrotizing Enterocolitis by Modulating Intestinal Injury and Repair." J Immunology 179:4808-4820.
Lee et al., 2006, "Homeostatic effects of TLR9 signaling in experimental colitis." Ann NY Acad Sci. 1072:351-5.
Lemaitre et al., "The dorsoventral regulatory gene cassette spatzle/Toll/cactus controls the potent antifungal response in Drosophila adults", Cell, 1996; 86:973-983.
Lin et al., "Oral probiotics reduce the incidence and severity of necrotizing enterocolitis in very low birth weight infants", Pediatrics, 2005; 115:1-4.
Lin et al., 2006, "Necrotising enterocolitis." Lancet 368:1271-1283.
Liu et al., "Changes in intestinal toll-like receptors and cytokines precede histological injury in a rat model of necrotizing enterocolitis", Am J Physiol Gastrointest Liver Physiol., 2009; 297:G442-G450.
Lotz et al., "Postnatal acquisition of endotoxin tolerance in intestinal epithelial cells", J Exp Med., 2006; 203:973-984.
Lu et al., "Polyunsaturated fatty acid supplementation alters proinflammatory gene expression and reduces the incidence of necrotizing enterocolitis in a neonatal rat model", Pediatr Res., 2007; 61:427-432.
Luig et al., "Epidemiology of necrotizing enterocolitis-PartI: Changing regional trends in extremely preterm infants over 14 years", J. Paediatr Child Health, 2005; 41(4):169-73.
Luig et al., "Epidemiology of necrotizing enterocolitis—PartI: Changing regional trends in extremely preterm infants over 14 years", J. Paediatr Child Health, 2005; 41(4):169-73.
Macagno et al., 2006, "A cyanobacterial LPS antagonist prevents endotoxin shock and.blocks sustained TLR4 stimulation required for cytokine expression." J. Exp. Med. 203(6):1481-1492.
Maeda et al., 2005, "Nod2 mutation in Crohn's disease potentiates NF-kappaB activity and IL- 1beta processing." Science 307:734-738. Erratum in Science. Apr. 29, 2005;308(5722):633.
Medzhitov et al., "A human homologue of the Drosophila Toll protein signals activation of adaptive immunity", Nature, 1997; 388:394-397.
Merck Manual website, Nov. 2007 by William J. Cochran, MD. Downloaded on Nov. 7, 2011 from <http://www.merckmanuals.com/professional/pediatrics/gastrointestinal_disorders_in_neonates_and_infants/necrotizing_enterocolitis.html>.
Michaelsson et al., "Regulation of T cell responses in the developing human fetus", J. Immunol., 2006; 176(10):5741-5748.
Milla et al., "Small intestinal motility patterns in the perinatal period", J Pediatr. Gastroenterol Nutr., 1983; 2:S141-S144.
Mizrahi et al., "Necrotizing enterocolitis in premature infants", J Pediatr., 1965; 66:697-705.
Moss et al., 2006, "Laparotomy versus peritoneal drainage for necrotizing enterocolitis and perforation." N. Engl. J. Med. 354:2225-2234.
Muguruma et al., "The central role of PAF in necrotizing enterocolitis development", Adv Exp Med Biol. 1997; 407:379-382.
Mullarkey et al., 2003, "Inhibition of endotoxin response by e5564, a novel Toll-like receptor 4-directed endotoxin antagonist." J Pharmacol Exp Ther. 304(3):1093-102.
Neal et al., "A critical role for TLR4 induction of autophagy in the regulation of enterocyte migration and the pathogenesis of necrotizing enterocolitis", J. Immunol., 2013; 190(7):3541-3551.
Neal et al., "Enterocyte TLR4 mediates phagocytosis and translocation of bacteria across the intestinal barrier." J Immunol. 176(5):3070-9 (2006).
Neu et al., 2005, "Intestinal innate immunity: how does it relate to the pathogenesis of necrotizing enterocolitis." Semin. Pediatr. Surg. 14: 137-144.
Neu, 1996, "Necrotizing enterocolitis: the search for a unifying pathogenic theory leading to prevention." Pediatr Clin North Am. 43(2):409-32.
Ng, 2001, "Necrotizing enterocolitis in the full-term neonate." J Paediatr Child Health. 37(1):1-4.
Noerr, "Current controversies in the understanding of necrotizing enterocolitis", Adv Neonatal Care, 2003; 3:107-120.
Obermeier et al. 2002, "CpG motifs of bacterial DNA exacerbate colitis of dextran sulfate sodium-treated mice." Eur J Immunol. Jul. 2002;32(7):2084-92.
Obermeier et al., "Contrasting activity of cytosin-guanosin dinucleotide oligonucleotides in mice with experimental colitis", Clin Exp Immunol., 134(2):217-224 (2003).
Ogura et al., "A frameshift mutation in NOD2 associated with susceptibility to Crohn's disease." Nature. 411(6837):603-6 (2001).
Otte et al., 2004, "Mechanisms of cross hyporesponsiveness to Toll-like receptor bacterial ligands in intestinal epithelial cells." Gastroenterology. 126(4):1054-70.
Panigrahi, "Necrotizing enterocolitis", Paediatr. Drugs, 2006; 8(3):151-165.
Parant et al., "Stimulation of Non-Specific Resistance to Infections by Synthetic Immunoregulatory Agents," Infection 12(3): 230-234, 1984.
Pierro, 2005, "The surgical management of necrotising enterocolitis." Early Hum Dev. 81(1):79-85.
Poltorak et al., 1998, "Defective LPS Signaling in C3H/HeJ and C57BL/10ScCr Mice: Mutations in Tlr4 Gene." Science 282: 2085-2088.
Prohinar et al., "Specific high affinity interactions of monomeric endotoxin protein complexes with Toll-like receptor 4 ectodomain." J Biol Chem. 282(2):1010-7. (2007).
Putta et al., 2006, "Novel oligodeoxynucleotide agonists of TLR9 containing N3-Me-dC or N1-Me-dG modifications." Nucleic Acids Res. 34(11):3231-8.
Qureshi et al., "Increased expression and function of integrins in enterocytes by endotoxin impairs epithelial restitution", Gastroenterology, 2005; 128:1012-1022.
Rachmelewitz et al., 2004, "Toll-like receptor 9 signaling mediates the anti-inflammatory effects of probiotics in murine experimental colitis." Gastroenterology. 126(2):520-8.
Rakoff-Nahoum et al., "Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis", Cell, 2004; 118:229-241.
Richardson, et al., "Nucleotide-binding Oligomerization Domain-2 Inhibits Toll Like Receptor-4 Signaling in the Intestinal Epithelium", Gastroenterology, 139(3):904-917 (2010).
Roach et al., "The evolution of vertebrate Toll-like receptors", PNAS, 2005; 102:9577-9582.
Rossignol et al., 2004, "Safety, pharmacokinetics, pharmacodynamics, and plasma lipoprotein distribution of eritoran (E5564) during continuous intravenous infusion into healthy volunteers." Antimicrob Agents Chemother. 48(9):3233-40.
Sartor, "Targeting enteric bacteria in treatment of inflammatory bowel diseases: why, how, and when," Current Opinion in Gastroenterology 2003, 19:358-365.
Shan et al., "Regulation of toll-like receptor 4-induced proasthmatic changes in airway smooth muscle function by opposing actions of ERK1/2 and p38 MAPK signaling", Am J. Physiol. Lung Cell Mol. Physiol., 291(3):L324-L333 (2006).
Sharma et al., 2007, "Neonatal gut barrier and multiple organ failure: role of endotoxin and proinflammatory cytokines in sepsis and necrotizing enterocolitis." J Pediatr Surg 42:454-461.
Shin et al., 2000, "Diminished epidermal growth factor levels in infants with necrotizing enterocolitis." J Pediatr Surg. 35(2):173-6; discussion 177.
Shindou et al., "Roles of cytosolic phospholipase A2 and platelet-activating factor receptor in the Ca-induced biosynthesis of PAF", Biochem Biophys Res Commun. 2000; 271:812-817.
Shuto et al., "Activation of NF-kappa B by nontypeable hemophilus influenzae is mediated by toll-like receptor 2-TAK1-dependent NIK-IKK alpha/beta-I kappa B alpha and MKK3/6-p38 MAP kinase signaling pathways in epithelial cells", PNAS, 98(15):8774-8779 (2001).
Sodhi, et al., "DNA Attenuates Enterocyte Toll-like Receptor 4-Mediated Intestinal Mucosal Injury After Remote Trauma", Am J Physiol Gastrointest Liver Physiol., 300:G862-G873 (2011).
Sodhi, et al., "Toll-like-receptor-4 Inhibits Enterocyte Proliferation via Impaired β-Catenin Signaling in Necrotizing Enterocolitis", Gastroenterology, 138(1):185-196 (2010).
Strober et al., 2006, "Signalling pathways and molecular interactions of NOD1 and NOD2." Nat Rev Immunol. 6:9-20.
Supplemental European Search Report for EP Application No. 08746070.5, dated May 25, 2011.
Svetlov et al., "Regulation of platelet-activating factor (PAF) biosynthesis via coenzyme A-independent transacylase in the macrophage cell line IC-21 stimulated with lipopolysaccharide", Biochim Biophys Acta, 1997; 1346:120-130.
Takeda et al., "Toll-like receptors in innate immunity." Int Immunol. 17(1):1-140, 2005.
Takeda et al., 2001, "Roles of Toll-like receptors in innate immune responses." Genes Cells 6:733-742.
Tatum et al., "The role of toll-like receptor 9 in an animal model of necrotizing entercolitis", Journal of Investigative Medicine, 58(2):436 (2010).
Thompson et al., "Necrotizing enterocolitis in newborns", Drugs, 2008; 68(9):1227-1238.
U.S. Appl. No. 12/104,816 (U.S. Pat. No. 8,188,058), filed Apr. 17, 2008 (May 29, 2012).
U.S. Appl. No. 12/104,816, Apr. 23, 2012 Issue Fee payment.
U.S. Appl. No. 12/104,816, Aug. 9, 2010 Restriction Requirement.
U.S. Appl. No. 12/104,816, Feb. 24, 2011 Response to Non-Final Office Action.
U.S. Appl. No. 12/104,816, Jan. 23, 2012 Notice of Allowance.
U.S. Appl. No. 12/104,816, May 10, 2011 Final Office Action.
U.S. Appl. No. 12/104,816, May 14, 2013 Certificate of Correction.
U.S. Appl. No. 12/104,816, Nov. 10, 2011 Amendment and Request for Continued Examination (RCE).
U.S. Appl. No. 12/104,816, Nov. 24, 2010 Non-Final Office Action.
U.S. Appl. No. 12/104,816, Sep. 15, 2010 Response to Restriction Requirement.
U.S. Appl. No. 13/068,553 (U.S. Pat. No. 8,518,903), filed May 13, 2011 (Aug. 27, 2013).
U.S. Appl. No. 13/068,553, Apr. 12, 2013 Response to Non-Final Office Action.
U.S. Appl. No. 13/068,553, Apr. 30, 2013 Notice of Allowance.
U.S. Appl. No. 13/068,553, Jan. 15, 2013 Non-Final Office Action.
U.S. Appl. No. 13/068,553, Jul. 25, 2013 Issue Fee payment.
U.S. Appl. No. 13/461,672 (U.S. Pat. No. 8,518,905), filed May 1, 2012 (Aug. 27, 2013).
U.S. Appl. No. 13/461,672, Apr. 12, 2013 Response to Non-Final Office Action.
U.S. Appl. No. 13/461,672, Apr. 29, 2013 Notice of Allowance.
U.S. Appl. No. 13/461,672, Jan. 14, 2013 Non-Final Office Action.
U.S. Appl. No. 13/461,672, Jul. 25, 2013 Issue Fee payment.
U.S. Appl. No. 13/848,809 (U.S. 2013/0281395), Mar. 22, 2013 (Oct. 24, 2013).
U.S. Appl. No. 13/848,809, Feb. 17, 2015 Notice of Allowance.
U.S. Appl. No. 13/848,809, Feb. 3, 2015 Request for Continued Examination (RCE).
U.S. Appl. No. 13/848,809, May 12, 2015 Issue Fee Payment.
U.S. Appl. No. 13/848,809, Nov. 10, 2014 Notice of Allowance.
U.S. Appl. No. 14/010,232 (U.S. 2015/0056217), filed Aug. 26, 2013 (Feb. 26, 2015).
U.S. Appl. No. 14/010,232, Aug. 10, 2016 Amendment and Request for Continued Examination (RCE).
U.S. Appl. No. 14/010,232, Feb. 27, 2015 Restriction Requirement Filed.
U.S. Appl. No. 14/010,232, Jun. 19, 2015 Non-Final Office Action.
U.S. Appl. No. 14/010,232, May 10, 2016 Final Office Action.
U.S. Appl. No. 14/010,232, May 27, 2015 Response to Restriction Requirement.
U.S. Appl. No. 14/010,232, Nov. 23, 2016 Issue Fee Payment.
U.S. Appl. No. 14/010,232, Sep. 21, 2015 Response to Non-Final Office Action.
U.S. Appl. No. 14/010,232, Sep. 28, 2016 Notice of Allowance.
U.S. Appl. No. 14/010,232.
U.S. Appl. No. 14/036,960 (U.S. 2014/0086982), Sep. 25, 2013 (Mar. 27, 2014).
U.S. Appl. No. 14/036,960, Dec. 21, 2016 Issue Fee Payment.
U.S. Appl. No. 14/036,960, Dec. 22, 2014 Response to Restriction Requirement.
U.S. Appl. No. 14/036,960, Jan. 12, 2015 Non-Final Office Action.
U.S. Appl. No. 14/036,960, Jan. 4, 2016 Response to Non-Final Office Action.
U.S. Appl. No. 14/036,960, Jul. 20, 2016 Response to Non-Final Office Action.
U.S. Appl. No. 14/036,960, Jul. 9, 2015 Response to Non-Final Office Action.
U.S. Appl. No. 14/036,960, Oct. 1, 2015 Non-Final Office Action.
U.S. Appl. No. 14/036,960, Oct. 26, 2016 Notice of Allowance.
U.S. Appl. No. 14/036,960, Sep. 23, 2014 Restriction Requirement Filed.
U.S. Appl. No. 14/036960, Apr. 20, 2016 Non-Final Office Action.
U.S. Appl. No. 14/717,349 (U.S. Pat. No. 9,532,999), filed May 20, 2015 (Jan. 3, 2017).
U.S. Appl. No. 14/717,349, Aug. 15, 2016 Response after Final Office Action.
U.S. Appl. No. 14/717,349, Aug. 29, 2016 Notice of Allowance.
U.S. Appl. No. 14/717,349, Dec. 10, 2015 Non-Final Office Action.
U.S. Appl. No. 14/717,349, Mar. 10, 2016 Response to Non-Final Office Action.
U.S. Appl. No. 14/717,349, May 26, 2016 Final Office Action.
U.S. Appl. No. 14/717,349, Nov. 22, 2016 Issue Fee Payment.
U.S. Appl. No. 15/383,625 (U.S. 2017/0095493), filed Dec. 19, 2016 (Apr. 6, 2017).
Uauy et al., 1991, "Necrotizing enterocolitis in very low birth weight infants: biodemographic and clinical correlates." National Institute of Child Health and Human Development Neonatal Research Network. J Pediatr 119:630-638.
University of Pittsburgh Department of Critical Care Medicine: Research-The Crisma Laboratory, pp. 1-11. Downloaded on Apr. 19, 2007 from http:/www.ccm.upmc.edu/research/res_crisma.htlm.
Van Heel et al., "Synergy between TLR9 and NOD2 innate immune responses is lost in genetic Chrohn's disease" GUT, British Medical Association, 54(11):1553-1557 (2005).
Verma et al., "Novel pharmacophores of connexin-43 based on the "RXP" series of Cx43-binding peptides", Circ. Res., 2009; 105(2):176-184.
Verthelyi et al., "Human peripheral blood cells differentially recognize and respond to two distinct CPG motifs." J Immunol. 166(4):2372-7 (2001).
Vink et al., 2002, "In vivo evidence for a role of toll-like receptor 4 in the development of intimal lesions." Circulation. 106(15):1985-90.
Wang et al., "NF-κB-mediated expression of MAPK phosphatase-1 is an early step in desensitization to TLR ligands in enterocytes", Mucosal Immunol., 2010; 3:523-534.
Wang et al., "Ubiquitin-editing enzyme A20 promotes tolerance to lipopolysaccharide in enterocytes", J Immunol., 2009; 183:1384-1392.
Warner et al., 2005, "Role of epidermal growth factor in the pathogenesis of neonatal necrotizing enterocolitis." Semin Pediatr Surg. 14(3):175-80.
Watanabe et al., "Muramyl dipeptide activation of nucleotide-binding oligomerization domain 2 protects mice from experimental colitis." J Clin Invest 118:545-559 (2008).
Wirtz et al., "Illuminating the role of type I IFNs in colitis." J Clin Invest. 115(3):586-8 (2005).
Wolfs et al., "Localization of the lipopolysaccharide recognition complex in the human healthy and inflamed premature and adult gut", Inflamm Bowel Dis., 2010; 16:68-75.
Worthen et al., "The priming of neutrophils by lipopolysaccharide for production of intracellular platelet-activating factor: potential role in mediation of enhanced superoxide secretion", J Immunol., 1988; 140:3553-3559.
Wynn et al., "The host response to sepsis and developmental impact", Pediatrics, 2010; 125:1031-1041.
Yang et al., "NOD2 transgenic mice exhibit enhanced MDP-mediated down-regulation of TLR2 responses and resistance to colitis induction." Gastroenterology 133:1510-1521 (2007).
Yang et al., 2005, "Role of Toll-like receptor 4/NF-kappaB pathway in monocyte-endothelial adhesion induced by low shear stress and ox-LDL." Biorheology. 42(3):225-36.
Yang et al., 2007, "NOD2 pathway activation by MDP or Mycobacterium tuberculosis infection involves the stable polyubiquitination of Rip2." J Biol Chem 282:36223-36229.
Zhai et al., "Cutting edge: TLR4 activation mediates liver ischemia/reperfusion inflammatory response via IFN regulatory factor 3-dependent MyD88-independent pathway", J. Immunol., 173(12):7115-7119 (2004).
Zheng et al., "Regulation of colonic epithelial repair in mice by Toll-like receptors and hyaluronic acid", Gastroenterology, 2009; 137:2041-2051.
Zhou et al., "Oral administration of plant-based rotavirus VP6 induces antigen-specific IgAs, IgGs and passive protection in mice" Vaccine, 28:6021-6027 (2010).
Zouali et al., "CARD15/NOD2 is Not a Predisposing Factor for Necrotizing Enterocolitis," Digestive Diseases and Sciences 50(9): 1684-1687, 2005.

Also Published As

Publication number Publication date
WO2012088425A2 (en) 2012-06-28
US20190070168A1 (en) 2019-03-07
WO2012088425A9 (en) 2012-10-11
US20130345154A1 (en) 2013-12-26

Similar Documents

Publication Publication Date Title
US20190070168A1 (en) Gap junction-enhancing agents for treatment of necrotizing enterocolitis and inflammatory bowel disease
JP4177224B2 (en) Use of ghrelin to treat low body weight and low body fat mass in individuals undergoing gastrectomy
CN103784937A (en) Methods for reducing CD36 expression
JP2007532495A (en) Use of secretagogues to treat ghrelin deficiency
ES2791778T3 (en) Mitigation of tissue damage and fibrosis through anti-LTBP4 antibodies
US20100152117A1 (en) Aequorin-Containing Compositions and Methods of Using Same
JP6031121B2 (en) Modulating ghrelin levels and ghrelin / non-acylated ghrelin ratio using unacylated ghrelin
JP2014504601A (en) Apolipoprotein AIV as an antidiabetic peptide
Han et al. Molecular cloning and functional expression of an LLC-PK1 cell taurine transporter that is adaptively regulated by taurine
EA011092B1 (en) Peptide, having stress-protective action, pharmaceutical composition based thereon and use thereof
RU2273645C9 (en) Obesity polypeptide (ob) (variants), its analogue (variants), and fused protein (variants), nucleic acid isolated molecule, dna molecule, cloning recombinant vector, expression recombinant vector, pharmaceutical composition, monoclonal and polyclonal antibody
WO2006030956A1 (en) NOVEL USE OF PrPP AND RECEPTOR THEREOF
US20100041598A1 (en) Use of Prolactin in the Prophylactic Treatment of Cancer
US9895410B2 (en) Methods for preventing and treating oral cancers
Talukder et al. Functional characterization, localization, and molecular identification of rabbit intestinal N-amino acid transporter
AU2005274377B2 (en) Method of screening for a carnitine transporter agonist or antagonist and its uses
US9925162B2 (en) Methods and compositions for inducing physiological hypertrophy
US9050296B2 (en) Methods for treating metabolic syndrome with Cthrc1 polypeptide
Moreau et al. Stanniocalcin-1 in the subfornical organ inhibits the dipsogenic response to angiotensin II
EP4046658A1 (en) Application of non-igf1r-binding substance in prevention and/or treatment of inflammatory diseases
US20060166860A1 (en) Zinc transporter compositions for the treatment of cardiovascular diseases
US20120142776A1 (en) Methods and compositions for inducing physiological hypertrophy
US20100081622A1 (en) Reduction of parathyroid hormone levels
EP1600165A1 (en) Medicinal use of mip-3alpha inhibitor and method of screening brain/nerve cell protective agent
US20240025949A1 (en) Beta-Arrestin Effectors and Compositions and Methods of Use Thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: UNIVERSITY OF PITTSBURGH - OF THE COMMONWEALTH SYS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:HACKAM, DAVID J.;REEL/FRAME:033772/0087

Effective date: 20130923

STCF Information on status: patent grant

Free format text: PATENTED CASE

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 4TH YR, SMALL ENTITY (ORIGINAL EVENT CODE: M2551); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

Year of fee payment: 4